Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Amino Acids ; 48(8): 2025-39, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26940723

RESUMO

Skeletal muscles require energy either at constant low (e.g., standing and posture) or immediate high rates (e.g., exercise). To fulfill these requirements, myocytes utilize the phosphocreatine (PCr)/creatine (Cr) system as a fast energy buffer and shuttle. We have generated mice lacking L-arginine:glycine amidino transferase (AGAT), the first enzyme of creatine biosynthesis. These AGAT(-/-) (d/d) mice are devoid of the PCr/Cr system and reveal severely altered oxidative phosphorylation. In addition, they exhibit complete resistance to diet-induced obesity, which is associated with a chronic activation of AMP-activated protein kinase in muscle and white adipose tissue. The underlying metabolic rearrangements have not yet been further analyzed. Here, we performed gene expression analysis in skeletal muscle and a serum amino acid profile of d/d mice revealing transcriptomic and metabolic alterations in pyruvate and glucose pathways. Differential pyruvate tolerance tests demonstrated preferential conversion of pyruvate to alanine, which was supported by increased protein levels of enzymes involved in pyruvate and alanine metabolism. Pyruvate tolerance tests suggested severely impaired hepatic gluconeogenesis despite increased availability of pyruvate and alanine. Furthermore, enzymes of serine production and one-carbon metabolism were significantly up-regulated in d/d mice, indicating increased de novo formation of one-carbon units from carbohydrate metabolism linked to NAD(P)H production. Besides the well-established function of the PCr/Cr system in energy metabolism, our transcriptomic and metabolic analyses suggest that it plays a pivotal role in systemic one-carbon metabolism, oxidation/reduction, and biosynthetic processes. Therefore, the PCr/Cr system is not only an energy buffer and shuttle, but also a crucial component involved in numerous systemic metabolic processes.


Assuntos
Amidinotransferases/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Deficiência Intelectual/metabolismo , Metaboloma , Obesidade/metabolismo , Fosforilação Oxidativa , Fosfocreatina/metabolismo , Distúrbios da Fala/metabolismo , Transcriptoma , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Amidinotransferases/genética , Amidinotransferases/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Fosfocreatina/genética , Distúrbios da Fala/genética , Distúrbios da Fala/patologia
2.
Front Med (Lausanne) ; 10: 1138644, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37077670

RESUMO

Metabolic causes such as altered bioenergetics and amino acid metabolism may play a major role in Long COVID. Renal-metabolic regulation is an integral part of these pathways but has not been systematically or routinely investigated in Long COVID. Here we discuss the biochemistry of renal tubular injury as it may contribute to Long COVID symptoms. We propose three potential mechanisms that could be involved in Long COVID namely creatine phosphate metabolism, un-reclaimed glomerular filtrate and COVID specific proximal tubule cells (PTC) injury-a tryptophan paradigm. This approach is intended to allow for improved diagnostics and therapy for the long-haul sufferers.

3.
Res Vet Sci ; 144: 175-180, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34823870

RESUMO

A striking increase in homoarginine concentrations, about more than 100-fold that observed in humans, was recently reported during pregnancy in a nutritionally induced model of intra-uterine growth restriction in ewes. To determine whether this phenomenon is at least partially related to the nutritional regimen, estrus synchronization, or analytical method, thirty-four one-year-old primiparous, non-synchronized, and well-fed Sarda breed ewes were exposed to fertile rams allowing those who came into estrus to naturally mate. Plasma arginine, homoarginine, asymmetric dimethylarginine, symmetric dimethylarginine, mono methylarginine, and citrulline concentrations were measured in each sample using LC-MS/MS. Homoarginine concentrations showed a 44-fold variation between the highest and the lowest values while the fluctuations of arginine and its analogues and metabolites were much smaller, between 1.1 and 1.6-fold. Repeated-measures correlation analysis showed a significant negative correlation between homoarginine/arginine and arginine/asymmetric dimethylarginine ratios (Rm = -0.40; P < 0.000001). Furthermore, median homoarginine concentrations significantly increased with the number of fetuses. The marked increase in homoarginine concentrations with advancing gestational age is genuine and independent of mating, feeding, diet, and hormone treatment. The higher homoarginine concentrations found in ewes bearing multiple fetuses suggest the presence of a physiological link between this arginine analog and energy metabolism in pregnancy that warrants further investigation.


Assuntos
Homoarginina , Espectrometria de Massas em Tandem , Animais , Cromatografia Líquida/veterinária , Feminino , Feto/metabolismo , Idade Gestacional , Homoarginina/metabolismo , Masculino , Gravidez , Ovinos , Espectrometria de Massas em Tandem/veterinária
4.
Front Nutr ; 9: 969702, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36017222

RESUMO

Organisms obtain creatine from their diet or by de novo synthesis via AGAT (L-arginine:glycine amidinotransferase) and GAMT (Guanidinoacetate N-methyltrasferase) in kidney and liver, respectively. AGAT also synthesizes homoarginine (hArg), low levels of which predict poor outcomes in human cardiovascular disease, while supplementation maintains contractility in murine heart failure. However, the expression pattern of AGAT has not been systematically studied in mouse tissues and nothing is known about potential feedback interactions between creatine and hArg. Herein, we show that C57BL/6J mice express AGAT and GAMT in kidney and liver respectively, whereas pancreas was the only organ to express appreciable levels of both enzymes, but no detectable transmembrane creatine transporter (Slc6A8). In contrast, kidney, left ventricle (LV), skeletal muscle and brown adipose tissue must rely on creatine transporter for uptake, since biosynthetic enzymes are not expressed. The effects of creatine and hArg supplementation were then tested in wild-type and AGAT knockout mice. Homoarginine did not alter creatine accumulation in plasma, LV or kidney, whereas in pancreas from AGAT KO, the addition of hArg resulted in higher levels of tissue creatine than creatine-supplementation alone (P < 0.05). AGAT protein expression in kidney was downregulated by creatine supplementation (P < 0.05), consistent with previous reports of end-product repression. For the first time, we show that hArg supplementation causes a similar down-regulation of AGAT protein (P < 0.05). These effects on AGAT were absent in the pancreas, suggesting organ specific mechanisms of regulation. These findings highlight the potential for interactions between creatine and hArg that may have implications for the use of dietary supplements and other therapeutic interventions.

5.
Front Physiol ; 9: 773, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30013483

RESUMO

Creatine serves as fast energy buffer in organs of high-energy demand such as brain and skeletal muscle. L-Arginine:glycine amidinotransferase (AGAT) and guanidinoacetate N-methyltransferase are responsible for endogenous creatine synthesis. Subsequent uptake into target organs like skeletal muscle, heart and brain is mediated by the creatine transporter (CT1, SLC6A8). Creatine deficiency syndromes are caused by defects of endogenous creatine synthesis or transport and are mainly characterized by intellectual disability, behavioral abnormalities, poorly developed muscle mass, and in some cases also muscle weakness. CT1-deficiency is estimated to be among the most common causes of X-linked intellectual disability and therefore the brain phenotype was the main focus of recent research. Unfortunately, very limited data concerning muscle creatine levels and functions are available from patients with CT1 deficiency. Furthermore, different CT1-deficient mouse models yielded conflicting results and detailed analyses of their muscular phenotype are lacking. Here, we report the generation of a novel CT1-deficient mouse model and characterized the effects of creatine depletion in skeletal muscle. HPLC-analysis showed strongly reduced total creatine levels in skeletal muscle and heart. MR-spectroscopy revealed an almost complete absence of phosphocreatine in skeletal muscle. Increased AGAT expression in skeletal muscle was not sufficient to compensate for insufficient creatine transport. CT1-deficient mice displayed profound impairment of skeletal muscle function and morphology (i.e., reduced strength, reduced endurance, and muscle atrophy). Furthermore, severely altered energy homeostasis was evident on magnetic resonance spectroscopy. Strongly reduced phosphocreatine resulted in decreased ATP/Pi levels despite an increased inorganic phosphate to ATP flux. Concerning glucose metabolism, we show increased glucose transporter type 4 expression in muscle and improved glucose clearance in CT1-deficient mice. These metabolic changes were associated with activation of AMP-activated protein kinase - a central regulator of energy homeostasis. In summary, creatine transporter deficiency resulted in a severe muscle weakness and atrophy despite different compensatory mechanisms.

6.
Reprod Sci ; 20(9): 1096-102, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23427185

RESUMO

We have previously reported that maternal creatine supplementation protects the neonate from hypoxic injury. Here, we investigated whether maternal creatine supplementation altered expression of the creatine synthesis enzymes (arginine:glycine amidinotransferase [AGAT], guanidinoaceteate methyltransferase [GAMT]) and the creatine transporter (solute carrier family 6 [neurotransmitter transporter, creatine] member 8: SLC6A8) in the term offspring. Pregnant spiny mice were fed a 5% creatine monohydrate diet from midgestation (day 20) to term (39 days). Placentas and neonatal kidney, liver, heart, and brain collected at 24 hours of age underwent quantitative polymerase chain reaction and Western blot analysis. Maternal creatine had no effect on the expression of AGAT and GAMT in neonatal kidney and liver, but mRNA expression of AGAT in brain tissues was significantly decreased in both male and female neonates born to mothers who were fed the creatine diet. SLC6A8 expression was not affected by maternal dietary creatine loading in any tissues. Maternal dietary creatine supplementation from midgestation in the spiny mouse did not alter the capacity for creatine synthesis or transport.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Creatinina/administração & dosagem , Creatinina/metabolismo , Suplementos Nutricionais , Fenômenos Fisiológicos da Nutrição Pré-Natal , Amidinotransferases/genética , Amidinotransferases/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Feminino , Idade Gestacional , Guanidinoacetato N-Metiltransferase/genética , Guanidinoacetato N-Metiltransferase/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Murinae , Miocárdio/metabolismo , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa