Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Biochem Biophys Res Commun ; 546: 59-64, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33571905

RESUMEN

Adenylate kinase2 (AK2) catalyzes trans-compartmental nucleotide exchange, but the functional implications of this mitochondrial intermembrane isoform is only partially understood. Here, transgenic AK2-/- null homozygosity was lethal early in embryo, indicating a mandatory role for intact AK2 in utero development. In the adult, conditional organ-specific ablation of AK2 precipitated abrupt heart failure with Krebs cycle and glycolytic metabolite buildup, suggesting a vital contribution to energy demanding cardiac performance. Depressed pump function recovered to pre-deletion levels overtime, suggestive of an adaptive response. Compensatory upregulation of phosphotransferase AK1, AK3, AK4 isozymes, creatine kinase isoforms, and hexokinase, along with remodeling of cell cycle/growth genes and mitochondrial ultrastructure supported organ rescue. Taken together, the requirement of AK2 in early embryonic stages, and the immediate collapse of heart performance in the AK2-deficient postnatal state underscore a primordial function of the AK2 isoform. Unsalvageable in embryo, loss of AK2 in the adult heart was recoverable, underscoring an AK2-integrated bioenergetics system with innate plasticity to maintain homeostasis on demand.


Asunto(s)
Adenilato Quinasa/metabolismo , Desarrollo Embrionario , Homeostasis , Miocardio/enzimología , Miocardio/metabolismo , Adaptación Fisiológica , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Animales , Ciclo del Ácido Cítrico , Pérdida del Embrión , Desarrollo Embrionario/genética , Metabolismo Energético , Femenino , Eliminación de Gen , Genes Esenciales/genética , Glucólisis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Homeostasis/genética , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Transgénicos
2.
J Am Soc Nephrol ; 31(5): 907-919, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32276962

RESUMEN

Growing evidence indicates that oxidative and endoplasmic reticular stress, which trigger changes in ion channels and inflammatory pathways that may undermine cellular homeostasis and survival, are critical determinants of injury in the diabetic kidney. Cells are normally able to mitigate these cellular stresses by maintaining high levels of autophagy, an intracellular lysosome-dependent degradative pathway that clears the cytoplasm of dysfunctional organelles. However, the capacity for autophagy in both podocytes and renal tubular cells is markedly impaired in type 2 diabetes, and this deficiency contributes importantly to the intensity of renal injury. The primary drivers of autophagy in states of nutrient and oxygen deprivation-sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), and hypoxia-inducible factors (HIF-1α and HIF-2α)-can exert renoprotective effects by promoting autophagic flux and by exerting direct effects on sodium transport and inflammasome activation. Type 2 diabetes is characterized by marked suppression of SIRT1 and AMPK, leading to a diminution in autophagic flux in glomerular podocytes and renal tubules and markedly increasing their susceptibility to renal injury. Importantly, because insulin acts to depress autophagic flux, these derangements in nutrient deprivation signaling are not ameliorated by antihyperglycemic drugs that enhance insulin secretion or signaling. Metformin is an established AMPK agonist that can promote autophagy, but its effects on the course of CKD have been demonstrated only in the experimental setting. In contrast, the effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors may be related primarily to enhanced SIRT1 and HIF-2α signaling; this can explain the effects of SGLT2 inhibitors to promote ketonemia and erythrocytosis and potentially underlies their actions to increase autophagy and mute inflammation in the diabetic kidney. These distinctions may contribute importantly to the consistent benefit of SGLT2 inhibitors to slow the deterioration in glomerular function and reduce the risk of ESKD in large-scale randomized clinical trials of patients with type 2 diabetes.


Asunto(s)
Autofagia/fisiología , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/etiología , Nutrientes/metabolismo , Oxígeno/metabolismo , Podocitos/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Adenilato Quinasa/deficiencia , Adenilato Quinasa/fisiología , Autofagia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/prevención & control , Progresión de la Enfermedad , Estrés del Retículo Endoplásmico , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Transporte Iónico/efectos de los fármacos , Túbulos Renales/citología , Túbulos Renales/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Mitocondrias/metabolismo , Estrés Oxidativo , Consumo de Oxígeno , Podocitos/patología , Circulación Renal/efectos de los fármacos , Transducción de Señal , Sirtuina 1/deficiencia , Sirtuina 1/fisiología , Sodio/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
3.
Sci Rep ; 9(1): 15739, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31673062

RESUMEN

Reticular dysgenesis is an autosomal recessive form of severe combined immunodeficiency (SCID) that usually manifests in newborns. It is a unique example of an immune deficiency that is linked to dysfunctional mitochondrial energy metabolism and caused by adenylate kinase 2 (AK2) deficiency. It is characterized by an early differentiation arrest in the myeloid lineage, impaired lymphoid maturation, and sensorineural hearing loss. In this study, a novel AK2 homozygous mutation, c.622 T > C [p.Ser208Pro], was identified in an Old Order Amish patient through whole exome sequencing. Functional studies showed that the patient's cells have no detectable AK2 protein, as well as low oxygen consumption rate (OCR), extracellular acidification rate (ECAR) and proton production rate (PPR). An increased production of reactive oxygen species, mitochondrial membrane permeability, and mitochondrial mass, and decreased ATP production, were also observed. The results confirm the pathogenicity of the AK2 mutation and demonstrate that reticular dysgenesis should be considered in Amish individuals presenting with immune deficiency. We also describe other pathophysiological aspects of AK2 deficiency not previously reported.


Asunto(s)
Adenilato Quinasa/genética , Leucopenia/diagnóstico , Mitocondrias/metabolismo , Inmunodeficiencia Combinada Grave/diagnóstico , Adenilato Quinasa/deficiencia , Médula Ósea/patología , Permeabilidad de la Membrana Celular , Preescolar , Metabolismo Energético , Fibroblastos/citología , Fibroblastos/metabolismo , Homocigoto , Humanos , Leucopenia/genética , Masculino , Membranas Mitocondriales/metabolismo , Consumo de Oxígeno , Linaje , Polimorfismo de Nucleótido Simple , Especies Reactivas de Oxígeno/metabolismo , Inmunodeficiencia Combinada Grave/genética , Secuenciación del Exoma
4.
J Clin Pathol ; 72(6): 393-398, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30918013

RESUMEN

Adenylate kinase (AK) deficiency is a rare erythroenzymopathy associated with hereditary nonspherocytic haemolytic anaemia along with mental/psychomotor retardation in few cases. Diagnosis of AK deficiency depends on the decreased level of enzyme activity in red cell and identification of a mutation in the AK1 gene. Until, only eight mutations causing AK deficiency have been reported in the literature. We are reporting two novel missense mutation (c.71A > G and c.413G > A) detected in the AK1 gene by next-generation sequencing (NGS) in a 6-year-old male child from India. Red cell AK enzyme activity was found to be 30% normal. We have screened a total of 32 family members of the patient and showed reduced red cell enzyme activity and confirm mutations by Sanger's sequencing. On the basis of Sanger sequencing, we suggest that the proband has inherited a mutation in AK1 gene exon 4 c.71A > G (p.Gln24Arg) from paternal family and exon 6 c.413G > A (p.Arg138His) from maternal family. Bioinformatics tools, such as SIFT, Polymorphism Phenotyping v.2, Mutation Taster, MutPred, also confirmed the deleterious effect of both the mutations. Molecular modelling suggests that the structural changes induced by p.Gln24Arg and p.Arg138His are pathogenic variants having a direct impact on the structural arrangement of the region close to the active site of the enzyme. In conclusion, NGS will be the best solution for diagnosis of very rare disorders leading to better management of the disease. This is the first report of the red cell AK deficiency from the Indian population.


Asunto(s)
Adenilato Quinasa/genética , Anemia Hemolítica Congénita no Esferocítica/genética , Eritrocitos/enzimología , Mutación Missense , Adenilato Quinasa/sangre , Adenilato Quinasa/química , Adenilato Quinasa/deficiencia , Adulto , Anemia Hemolítica Congénita no Esferocítica/sangre , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia Hemolítica Congénita no Esferocítica/enzimología , Niño , Análisis Mutacional de ADN/métodos , Femenino , Predisposición Genética a la Enfermedad , Herencia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , India , Masculino , Modelos Moleculares , Linaje , Fenotipo , Conformación Proteica , Relación Estructura-Actividad
5.
Br J Haematol ; 180(5): 644-653, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29270983

RESUMEN

Reticular Dysgenesis is a rare immunodeficiency which is clinically characterized by the combination of Severe Combined Immunodeficiency (SCID) with agranulocytosis and sensorineural deafness. Mutations in the gene encoding adenylate kinase 2 (AK2) were identified to cause this phenotype. In this review, we will demonstrate important clinical differences between reticular dysgenesis and other SCID entities and summarize recent concepts in the understanding of the pathophysiology of the disease and the management strategies for this difficult condition.


Asunto(s)
Leucopenia/genética , Leucopenia/terapia , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , Adenilato Quinasa/química , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Animales , Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/genética , Humanos , Síndromes de Inmunodeficiencia/genética , Leucopenia/diagnóstico , Mutación/genética , Inmunodeficiencia Combinada Grave/diagnóstico
7.
Cell Death Dis ; 6: e1856, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26270350

RESUMEN

Reticular dysgenesis is a human severe combined immunodeficiency that is primarily characterized by profound neutropenia and lymphopenia. The condition is caused by mutations in the adenylate kinase 2 (AK2) gene, resulting in the loss of mitochondrial AK2 protein expression. AK2 regulates the homeostasis of mitochondrial adenine nucleotides (ADP, ATP and AMP) by catalyzing the transfer of high-energy phosphate. Our present results demonstrate that AK2-knocked-down progenitor cells have poor proliferative and survival capacities and are blocked in their differentiation toward lymphoid and granulocyte lineages. We also observed that AK2 deficiency impaired mitochondrial function in general and oxidative phosphorylation in particular - showing that AK2 is critical in the control of energy metabolism. Loss of AK2 disrupts this regulation and leads to a profound block in lymphoid and myeloid cell differentiation.


Asunto(s)
Adenilato Quinasa/genética , Leucopenia/genética , Linfocitos/enzimología , Mitocondrias/genética , Neutrófilos/enzimología , Inmunodeficiencia Combinada Grave/genética , Células Madre/enzimología , Nucleótidos de Adenina/metabolismo , Adenilato Quinasa/deficiencia , Antígenos CD34/genética , Antígenos CD34/metabolismo , Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HL-60 , Humanos , Leucopenia/enzimología , Leucopenia/patología , Linfocitos/patología , Mitocondrias/enzimología , Mitocondrias/patología , Mutación , Neutrófilos/patología , Fosforilación Oxidativa , Cultivo Primario de Células , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/patología , Células Madre/patología
8.
Metab Brain Dis ; 30(6): 1369-77, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26152932

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective loss of motoneurons. While the principal cause of the disease remains so far unknown, the onset and progression of the pathology are increasingly associated with alterations in the control of cell metabolism. On the basis of the well-known key roles of 5'-adenosine monophosphate-activated protein kinase (AMPK) in sensing and regulating the intracellular energy status, we hypothesized that mice with a genetic deletion of AMPK would develop locomotor abnormalities that bear similarity with those detected in the very early disease stage of mice carrying the ALS-associated mutated gene hSOD1(G93A). Using an automated gait analysis system (CatWalk), we here show that hSOD1(G93A) mice and age-matched mice lacking the neuronal and skeletal muscle predominant α2 catalytic subunit of AMPK showed an altered gait, clearly different from wild type control mice. Double mutant mice lacking AMPK α2 and carrying hSOD1(G93A) showed the same early gait abnormalities as hSOD1(G93A) mice over an age span of 8 to 16 weeks. Taken together, these data support the concept that altered AMPK function and associated bioenergetic abnormalities could constitute an important component in the early pathogenesis of ALS. Therapeutic interventions acting on metabolic pathways could prove beneficial on early locomotor deficits, which are sensitively detectable in rodent models using the CatWalk system.


Asunto(s)
Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/psicología , Trastornos Neurológicos de la Marcha/metabolismo , Trastornos Neurológicos de la Marcha/psicología , Envejecimiento/psicología , Animales , Progresión de la Enfermedad , Metabolismo Energético/genética , Trastornos Neurológicos de la Marcha/etiología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
9.
J Exp Med ; 212(8): 1185-202, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26150473

RESUMEN

Adenylate kinases (AKs) are phosphotransferases that regulate the cellular adenine nucleotide composition and play a critical role in the energy homeostasis of all tissues. The AK2 isoenzyme is expressed in the mitochondrial intermembrane space and is mutated in reticular dysgenesis (RD), a rare form of severe combined immunodeficiency (SCID) in humans. RD is characterized by a maturation arrest in the myeloid and lymphoid lineages, leading to early onset, recurrent, and overwhelming infections. To gain insight into the pathophysiology of RD, we studied the effects of AK2 deficiency using the zebrafish model and induced pluripotent stem cells (iPSCs) derived from fibroblasts of an RD patient. In zebrafish, Ak2 deficiency affected hematopoietic stem and progenitor cell (HSPC) development with increased oxidative stress and apoptosis. AK2-deficient iPSCs recapitulated the characteristic myeloid maturation arrest at the promyelocyte stage and demonstrated an increased AMP/ADP ratio, indicative of an energy-depleted adenine nucleotide profile. Antioxidant treatment rescued the hematopoietic phenotypes in vivo in ak2 mutant zebrafish and restored differentiation of AK2-deficient iPSCs into mature granulocytes. Our results link hematopoietic cell fate in AK2 deficiency to cellular energy depletion and increased oxidative stress. This points to the potential use of antioxidants as a supportive therapeutic modality for patients with RD.


Asunto(s)
Adenilato Quinasa/metabolismo , Células Madre Hematopoyéticas/fisiología , Leucopenia/enzimología , Leucopenia/fisiopatología , Estrés Oxidativo/fisiología , Células Madre Pluripotentes/fisiología , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/fisiopatología , Naranja de Acridina , Adenilato Quinasa/deficiencia , Animales , Antioxidantes/farmacología , Apoptosis/fisiología , Compuestos Azo , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Biología Computacional , Cartilla de ADN/genética , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Datos de Secuencia Molecular , Naftalenos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Pez Cebra
10.
J Med Invest ; 61(1-2): 137-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705759

RESUMEN

Adenylate kinase isozyme 2 (AK2) is located in mitochondrial intermembrane space and regulates energy metabolism by reversibly converting ATP and AMP to 2 ADPs. We previously demonstrated that disruption of the Drosophila melanogaster AK2 gene (Dak2) resulted in growth arrest during the larval stage and subsequent death. Two other groups found that human AK2 mutations cause reticular dysgenesis, a form of severe combined immunodeficiency (SCID) that is associated with severe hematopoietic defects and sensorineural deafness. However, the mechanisms underlying differential outcomes of AK2 deficiency in Drosophila and human systems remain unknown. In this study, effects of tissue-specific inactivation of the Dak2 gene on Drosophila development were analyzed using RNAi-mediated gene knockdown. In addition, to investigate the roles of AK2 in the regulation of gene expression during development, microarray analysis was performed using RNA from first and second instar larvae of Dak2-deficient mutant and wild-type D. melanogaster. Knockdown of Dak2 in all germ layers caused cessation of growth and subsequent death of flies. Microarray analysis revealed that Dak2 deficiency downregulates various genes, particularly those involved in the proteasomal function and in mitochondrial translation machinery. These data indicate that adenine nucleotide interconversion by Dak2 is crucial for developmental processes of Drosophila melanogaster.


Asunto(s)
Adenilato Quinasa/deficiencia , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Genes del Desarrollo/fisiología , Larva/genética , Adenilato Quinasa/genética , Animales , Regulación hacia Abajo/fisiología , Técnicas de Inactivación de Genes , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Supervivencia
11.
Arch Biochem Biophys ; 533(1-2): 11-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23474458

RESUMEN

Adenylate kinase isozyme 4 (AK4) belongs to a family of nucleotide monophosphate kinases involved in energy metabolism. Recently, AK4 was reported to play a role in protection from stress: In HEK293 cells, hypoxia increases AK4 expression but does not affect proliferation or viability, while RNA interference (RNAi) directed against AK4 inhibits proliferation and promotes death. By contrast, we show here that HepG2 cells showed much higher AK4 levels, which decreased under hypoxia along with markedly reduced cell proliferation and increased cell death. Nevertheless, RNAi directed against AK4 inhibited cell proliferation and caused death in both cell types, although cell cycle parameters were affected only in HepG2 cells. Hence reductions of AK4 levels were always associated with cell death. These results extend the notion of a stress-protective function of AK4 to a novel physiological context and show that AK4-mediated stress protection is not limited to one particular death scenario. Our data also allow the hypothesis that the different basal AK4 levels reflect different basal stress levels, causing alternative responses to additional stress.


Asunto(s)
Adenilato Quinasa/genética , Regulación Enzimológica de la Expresión Génica , Estrés Fisiológico/genética , Adenilato Quinasa/deficiencia , Adenilato Quinasa/metabolismo , Muerte Celular/genética , Hipoxia de la Célula/genética , Proliferación Celular , Células HEK293 , Células Hep G2 , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , Regulación hacia Arriba
12.
Physiol Genomics ; 44(7): 386-402, 2012 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-22234996

RESUMEN

Next-generation screening of disease-related metabolomic phenotypes requires monitoring of both metabolite levels and turnover rates. Stable isotope (18)O-assisted (31)P nuclear magnetic resonance (NMR) and mass spectrometry uniquely allows simultaneous measurement of phosphometabolite levels and turnover rates in tissue and blood samples. The (18)O labeling procedure is based on the incorporation of one (18)O into P(i) from [(18)O]H(2)O with each act of ATP hydrolysis and the distribution of (18)O-labeled phosphoryls among phosphate-carrying molecules. This enables simultaneous recording of ATP synthesis and utilization, phosphotransfer fluxes through adenylate kinase, creatine kinase, and glycolytic pathways, as well as mitochondrial substrate shuttle, urea and Krebs cycle activity, glycogen turnover, and intracellular energetic communication. Application of expanded (18)O-labeling procedures has revealed significant differences in the dynamics of G-6-P[(18)O] (glycolysis), G-3-P[(18)O] (substrate shuttle), and G-1-P[(18)O] (glycogenolysis) between human and rat atrial myocardium. In human atria, the turnover of G-3-P[(18)O], which defects are associated with the sudden death syndrome, was significantly higher indicating a greater importance of substrate shuttling to mitochondria. Phosphometabolomic profiling of transgenic hearts deficient in adenylate kinase (AK1-/-), which altered levels and mutations are associated to human diseases, revealed a stress-induced shift in metabolomic profile with increased CrP[(18)O] and decreased G-1-P[(18)O] metabolic dynamics. The metabolomic profile of creatine kinase M-CK/ScCKmit-/--deficient hearts is characterized by a higher G-6-[(18)O]P turnover rate, G-6-P levels, glycolytic capacity, γ/ß-phosphoryl of GTP[(18)O] turnover, as well as ß-[(18)O]ATP and ß-[(18)O]ADP turnover, indicating altered glycolytic, guanine nucleotide, and adenylate kinase metabolic flux. Thus, (18)O-assisted gas chromatography-mass spectrometry and (31)P NMR provide a suitable platform for dynamic phosphometabolomic profiling of the cellular energetic system enabling prediction and diagnosis of metabolic diseases states.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Metabolómica/métodos , Miocardio/metabolismo , Adenilato Quinasa/deficiencia , Animales , Creatina Quinasa/metabolismo , Atrios Cardíacos/metabolismo , Humanos , Técnicas In Vitro , Marcaje Isotópico , Ratones , Ratones Transgénicos , Modelos Animales , Miocardio/enzimología , Isótopos de Oxígeno , Isótopos de Fósforo , Fosforilación , Ratas , Estrés Fisiológico
13.
Artículo en Inglés | MEDLINE | ID: mdl-20008220

RESUMEN

Congenital neutropenia comprises a variety of genetically heterogeneous phenotypic traits. Molecular elucidation of the underlying genetic defects has yielded important insights into the physiology of neutrophil differentiation and function. Non-syndromic variants of congenital neutropenia are caused by mutations in ELA2, HAX1, GFI1, or WAS. Syndromic variants of congenital neutropenia may be due to mutations in genes controlling glucose metabolism (SLC37A4, G6PC3) or lysosomal function (LYST, RAB27A, ROBLD3/p14, AP3B1, VPS13B). Furthermore, defects in genes encoding ribosomal proteins (SBDS, RMRP) and mitochondrial proteins (AK2, TAZ) are associated with congenital neutropenia syndromes. Despite remarkable progress in the field, many patients with congenital neutropenia cannot yet definitively be classified by genetic terms. This review addresses diagnostic and therapeutic aspects of congenital neutropenia and covers recent molecular and pathophysiological insights of selected congenital neutropenia syndromes.


Asunto(s)
Neutropenia/congénito , Proteínas Adaptadoras Transductoras de Señales , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Metabolismo de los Hidratos de Carbono/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Heterogeneidad Genética , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Lisosomas , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Neutropenia/tratamiento farmacológico , Neutropenia/genética , Neutropenia/fisiopatología , Neutropenia/cirugía , Proteínas/genética , Proteínas Ribosómicas/deficiencia , Proteínas Ribosómicas/genética , Síndrome , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Trasplante Homólogo , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética
14.
Am J Physiol Endocrinol Metab ; 296(5): E1013-21, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19276392

RESUMEN

The present study was performed to investigate the involvement of SNARK in physical activity levels in mice. To examine the acute effect of SNARK deficiency on voluntary running, Snark-deficient mice (Snark(+/-): n = 16) and their wild-type counterparts (Snark(+/+): n = 16) were assigned to sedentary or exercise (1 wk voluntary wheel running) groups. In addition, to clarify the differences in voluntary running activity and its effect between genotypes, mice (Snark(+/+): n = 16; Snark(+/-): n = 16) were also kept in individual cages with/without a running wheel for 5 mo. Unexpectedly, in both voluntary running experiments, running distances were increased in Snark(+/-) mice compared with Snark(+/+) mice. Under sedentary conditions, body and white adipose tissue weights were increased significantly in Snark(+/-) mice. However, no significant differences were observed between the two genotypes under exercise conditions, and the values were significantly less than those under sedentary conditions in the long-term experiment. In the short-term experiment, serum interleukin-6 level in exercised Snark(+/+) mice was the same as that in sedentary Snark(+/+) mice, whereas that in sedentary Snark(+/-) mice was significantly lower than in the other groups. In contrast, serum leptin level was reduced significantly in exercised Snark(+/-) mice compared with sedentary Snark(+/-) mice. The results of this study demonstrated that exposure to an environment that allows voluntary exercise promotes increased running activity and prevents obesity in Snark-deficient mice.


Asunto(s)
Adenilato Quinasa/deficiencia , Obesidad/enzimología , Condicionamiento Físico Animal/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Carrera/fisiología , Adenilato Quinasa/metabolismo , Animales , Temperatura Corporal/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Femenino , Glucógeno/metabolismo , Histocitoquímica/veterinaria , Interleucina-6/sangre , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Obesidad/sangre , Obesidad/prevención & control , Tamaño de los Órganos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo
15.
NMR Biomed ; 22(5): 523-31, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19156695

RESUMEN

We developed a new dedicated measurement protocol for dynamic (31)P MRS analysis in contracting calf muscles of the mouse, using minimally invasive assessment of the contractile force combined with the acquisition of spectroscopic data gated to muscle contraction and determination of phosphocreatine (PCr) recovery rate and ATP contractile cost. This protocol was applied in a comparative study of six wild type (WT) mice and six mice deficient in cytosolic creatine kinase and adenylate kinase isoform 1 (MAK(-/-) mice) using 70 repeated tetanic contractions at two contractions per minute. Force levels during single contractions, and metabolite levels and tissue pH during resting conditions were similar in muscles of MAK(-/-) and WT mice. Strikingly, muscle relaxation after contraction was significantly delayed in MAK(-/-) mice, but during repeated contractions, the decrease in the force was similar in both mouse types. Gated data acquisition showed a negligible PCr breakdown in MAK(-/-) immediately after contraction, without a concomitant decrease in ATP or tissue pH. This protocol enabled the determination of rapid PCr changes that would otherwise go unnoticed due to intrinsic low signal-to-noise ratio (SNR) in mouse skeletal muscles combined with an assessment of the PCr recovery rate. Our results suggest that MAK(-/-) mice use alternative energy sources to maintain force during repeated contractions when PCr breakdown is reduced. Furthermore, the absence of large increases in adenosine diphosphate (ADP) or differences in force compared to WT mice in our low-intensity protocol indicate that creatine kinase (CK) and adenylate kinase (AK) are especially important in facilitating energy metabolism during very high energy demands.


Asunto(s)
Adenilato Quinasa/deficiencia , Creatina Quinasa/deficiencia , Citosol/enzimología , Espectroscopía de Resonancia Magnética/métodos , Contracción Muscular/fisiología , Fosfocreatina/metabolismo , Adenilato Quinasa/metabolismo , Animales , Fenómenos Biomecánicos , Creatina Quinasa/metabolismo , Masculino , Ratones , Isótopos de Fósforo
16.
Nat Genet ; 41(1): 106-11, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19043416

RESUMEN

Reticular dysgenesis is an autosomal recessive form of human severe combined immunodeficiency characterized by an early differentiation arrest in the myeloid lineage and impaired lymphoid maturation. In addition, affected newborns have bilateral sensorineural deafness. Here we identify biallelic mutations in AK2 (adenylate kinase 2) in seven individuals affected with reticular dysgenesis. These mutations result in absent or strongly decreased protein expression. We then demonstrate that restoration of AK2 expression in the bone marrow cells of individuals with reticular dysgenesis overcomes the neutrophil differentiation arrest, underlining its specific requirement in the development of a restricted set of hematopoietic lineages. Last, we establish that AK2 is specifically expressed in the stria vascularis region of the inner ear, which provides an explanation of the sensorineural deafness in these individuals. These results identify a previously unknown mechanism involved in regulation of hematopoietic cell differentiation and in one of the most severe human immunodeficiency syndromes.


Asunto(s)
Adenilato Quinasa/deficiencia , Pérdida Auditiva Sensorineural/complicaciones , Pérdida Auditiva Sensorineural/enzimología , Sistema Hematopoyético/patología , Isoenzimas/deficiencia , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Animales , Diferenciación Celular , Línea Celular , Oído Interno/enzimología , Oído Interno/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Pérdida Auditiva Sensorineural/genética , Humanos , Recién Nacido , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Mutación/genética , Neutrófilos/patología , Linaje , Transporte de Proteínas , Inmunodeficiencia Combinada Grave/complicaciones , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología
18.
Acta Physiol (Oxf) ; 192(3): 413-20, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17973952

RESUMEN

AIM: Recently it was reported that adenylate kinase-1 knockout mice (AK(-/-)) exhibit elevated rates of glucose uptake following repeated contractions and hypoxia, but the mechanism was not investigated. The purpose of the present study was to measure the changes in glucose transport and AMP-activated protein kinase (AMPK) phosphorylation/activity following repeated contractions in isolated muscles from AK(-/-) mice. METHODS: Extensor digitorum longus muscles underwent an intense stimulation protocol that decreased force to less than 10% of initial by the end of 10 min. Glucose uptake was measured with 2-deoxy-D-[1,2-(3)H]glucose. RESULTS: Muscle glucose uptake in the basal state was identical between control and AK(-/-) mice and increased twofold in both groups during contraction. The general antioxidant: N-acetylcysteine, decreased contraction-mediated glucose uptake by 30% in both groups. AMPK activity and phosphorylation were similar in the two groups in the basal state and, surprisingly, after contraction as well (approximately threefold increase). Both groups exhibited marked decreases in adenosine triphosphate following contraction (60-70% depletion), which coincided with stoichiometric increases in the content of inosine monophosphate, an indirect marker of AMP production. Adenylate kinase activity averaged 2081 +/- 106 micromol min(-1) (g dry wt)(-1) for control and 37 +/- 10 for AK(-/-) muscles; the activity in the AK(-/-) muscle is likely accounted for by isoforms other than AK1. CONCLUSION: In conclusion, AK(-/-) mice have a normal capacity for contraction-mediated glucose uptake. This appears to occur via increases in AMP and reactive oxygen species that result in the activation of AMPK.


Asunto(s)
Adenilato Quinasa/metabolismo , Glucosa/metabolismo , Isoenzimas/metabolismo , Complejos Multienzimáticos/biosíntesis , Contracción Muscular/fisiología , Músculo Esquelético/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Quinasas Activadas por AMP , Acetilcisteína/farmacología , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Animales , Antioxidantes/farmacología , Transporte Biológico , Estimulación Eléctrica , Silenciador del Gen , Técnicas In Vitro , Isoenzimas/deficiencia , Isoenzimas/genética , Ratones , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Rápida/enzimología , Fibras Musculares de Contracción Rápida/fisiología , Relajación Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Fosforilación
19.
J Biol Chem ; 282(43): 31366-72, 2007 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17704060

RESUMEN

Matching blood flow to myocardial energy demand is vital for heart performance and recovery following ischemia. The molecular mechanisms responsible for transduction of myocardial energetic signals into reactive vasodilatation are, however, elusive. Adenylate kinase, associated with AMP signaling, is a sensitive reporter of the cellular energy state, yet the contribution of this phosphotransfer system in coupling myocardial metabolism with coronary flow has not been explored. Here, knock out of the major adenylate kinase isoform, AK1, disrupted the synchrony between inorganic phosphate P(i) turnover at ATP-consuming sites and gamma-ATP exchange at ATP synthesis sites, as revealed by (18)O-assisted (31)P NMR. This reduced energetic signal communication in the post-ischemic heart. AK1 gene deletion blunted vascular adenylate kinase phosphotransfer, compromised the contractility-coronary flow relationship, and precipitated inadequate coronary reflow following ischemia-reperfusion. Deficit in adenylate kinase activity abrogated AMP signal generation and reduced the vascular adenylate kinase/creatine kinase activity ratio essential for the response of metabolic sensors. The sarcolemma-associated splice variant AK1beta facilitated adenosine production, a function lost in the absence of adenylate kinase activity. Adenosine treatment bypassed AK1 deficiency and restored post-ischemic flow to wild-type levels, achieving phenotype rescue. AK1 phosphotransfer thus transduces stress signals into adequate vascular response, providing linkage between cell bioenergetics and coronary flow.


Asunto(s)
Adenilato Quinasa/fisiología , Isoenzimas/genética , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Transducción de Señal , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Animales , Creatina Quinasa/metabolismo , Metabolismo Energético , Eliminación de Gen , Corazón/fisiopatología , Isoenzimas/deficiencia , Isoenzimas/metabolismo , Isoenzimas/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Contracción Miocárdica , Isquemia Miocárdica/fisiopatología , Reperfusión Miocárdica , Miocardio/citología , Miocitos Cardíacos/metabolismo , Resonancia Magnética Nuclear Biomolecular , Isótopos de Oxígeno/metabolismo , Fosfoproteínas/metabolismo , Isótopos de Fósforo/metabolismo
20.
Exp Hematol ; 35(8): 1182-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17662886

RESUMEN

OBJECTIVE: Red cell adenylate kinase (AK) deficiency is a rare hereditary erythroenzymopathy associated with moderate to severe nonspherocytic hemolytic anemia and, in some cases, with mental retardation and psychomotor impairment. To date, diagnosis of AK deficiency depends upon demonstration of low enzyme activity in red blood cells and detection of mutations in AK1 gene. To investigate the molecular bases of the AK deficiency, we characterized five variants of AK1 isoenzyme-bearing mutations (118G>A, 190G>A, 382C>T, 418-420del, and 491A>G) found in AK-deficient patients with chronic hemolytic anemia. MATERIALS AND METHODS: The complete AK1 cDNA was obtained by standard procedures and using as template the reticulocyte RNA. The cDNA was cloned in a plasmid vector and the enzyme was expressed in Escherichia coli BL21(DE3)pLysS, and purified by standard protocols to homogeneity. DNA mutants bearing point mutations were obtained from the cloned wild-type cDNA using standard methods of site-directed mutagenesis, whereas the DNA mutant with deletion of codon 140 was obtained by a two-step method. RESULTS: Four mutant enzymes (Gly40Arg, Gly64Arg, Arg128Trp, Asp140del) were severely affected in activity, displaying a catalytic efficiency of four orders of magnitude lower than the wild-type; one (Tyr164Cys) was grossly perturbed in protein stability. CONCLUSIONS: The altered properties displayed by the mutant enzymes support the cause-effect relationship between AK1 mutations and hemolytic anemia.


Asunto(s)
Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Anemia Hemolítica Congénita no Esferocítica/enzimología , Anemia Hemolítica Congénita no Esferocítica/genética , Eritrocitos/enzimología , Mutación , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/sangre , Adenilato Quinasa/química , Anemia Hemolítica Congénita no Esferocítica/sangre , Dicroismo Circular , Estabilidad de Enzimas , Escherichia coli/genética , Mutación del Sistema de Lectura , Humanos , Cinética , Modelos Moleculares , Mutación Missense , Reacción en Cadena de la Polimerasa , Conformación Proteica , ARN/genética , ARN/aislamiento & purificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/química , Mapeo Restrictivo , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...