Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Nutrients ; 14(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35057527

RESUMO

Carnitine deficiency is prevalent in patients undergoing hemodialysis, and it could result in lowered muscle strength. So far, the effect of treatment with levocarnitine on lower limb muscle strength has not been well described. This observational study examined the association between treatment with levocarnitine with the change in knee extensor strength (KES) in hemodialysis patients. Eligible patients were selected from the participants enrolled in a prospective cohort study for whom muscle strength was measured annually. We identified 104 eligible patients for this analysis. During the one-year period between 2014 to 2015, 67 patients were treated with intravenous levocarnitine (1000 mg per shot, thrice weekly), whereas 37 patients were not. The change in KES was significantly higher (p = 0.01) in the carnitine group [0.02 (0.01-0.04) kgf/kg] as compared to the non-carnitine group [-0.02 (-0.04 to 0.01) kgf/kg]. Multivariable-adjusted regression analysis showed the positive association between the change in KES and the treatment with levocarnitine remained significant after adjustment for the baseline KES and other potential confounders. Thus, treatment with intravenous levocarnitine was independently and positively associated with the change in KES among hemodialysis patients. Further clinical trials are needed to provide more solid evidence.


Assuntos
Cardiomiopatias/terapia , Carnitina/administração & dosagem , Carnitina/deficiência , Hiperamonemia/terapia , Força Muscular/efeitos dos fármacos , Doenças Musculares/terapia , Diálise Renal/efeitos adversos , Administração Intravenosa , Idoso , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Feminino , Humanos , Hiperamonemia/etiologia , Hiperamonemia/fisiopatologia , Joelho/fisiopatologia , Masculino , Pessoa de Meia-Idade , Doenças Musculares/etiologia , Doenças Musculares/fisiopatologia , Estudos Prospectivos , Análise de Regressão , Resultado do Tratamento
2.
Anal Biochem ; 636: 114437, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34715068

RESUMO

Hepatic encephalopathy and depression share a number of clinical features, such as cognitive impairment and psychomotor retardation, and are highly prevalent in patients with chronic liver disease. Both conditions signify a poor prognosis, carry an increased mortality and are major determinants of reduced health related quality of life. The pathophysiology of hepatic encephalopathy is complex. Whilst cerebral accumulation of ammonia is well-recognised as being central to the development of hepatic encephalopathy, systemic inflammation, which acts in synergy with hyperammonaemia, is emerging as a key driver in its development. The pro-inflammatory state is also widely documented in depression, and peripheral to brain communication occurs resulting in central inflammation, behavioural changes and depressive symptoms. Gut dysbiosis, with a similar reduction in beneficial bacteria, increase in pathogens and decreased bacterial diversity, has been observed in both hepatic encephalopathy and depression, and it may be that the resultant increased bacterial translocation causes their shared inflammatory pathophysiology. Whilst the literature on a positive association between hepatic encephalopathy and depression in cirrhosis remains to be substantiated, there is evolving evidence that treatment with psychobiotics may be of dual benefit, improving cognition and mood in cirrhosis.


Assuntos
Transtornos Cognitivos , Depressão , Encefalopatia Hepática , Hiperamonemia , Cirrose Hepática , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Doença Crônica , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/fisiopatologia , Depressão/etiologia , Depressão/metabolismo , Depressão/fisiopatologia , Encefalopatia Hepática/etiologia , Encefalopatia Hepática/metabolismo , Encefalopatia Hepática/fisiopatologia , Humanos , Hiperamonemia/etiologia , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Cirrose Hepática/complicações , Cirrose Hepática/metabolismo , Cirrose Hepática/fisiopatologia , Qualidade de Vida
3.
Anal Biochem ; 636: 114343, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34637785

RESUMO

Urea cycle disorders (UCD) are inherited diseases resulting from deficiency in one of six enzymes or two carriers that are required to remove ammonia from the body. UCD may be associated with neurological damage encompassing a spectrum from asymptomatic/mild to severe encephalopathy, which results in most cases from Hyperammonemia (HA) and elevation of other neurotoxic intermediates of metabolism. Electroencephalography (EEG), Magnetic resonance imaging (MRI) and Proton Magnetic resonance spectroscopy (MRS) are noninvasive measures of brain function and structure that can be used during HA to guide management and provide prognostic information, in addition to being research tools to understand the pathophysiology of UCD associated brain injury. The Urea Cycle Rare disorders Consortium (UCDC) has been invested in research to understand the immediate and downstream effects of hyperammonemia (HA) on brain using electroencephalogram (EEG) and multimodal brain MRI to establish early patterns of brain injury and to track recovery and prognosis. This review highlights the evolving knowledge about the impact of UCD and HA in particular on neurological injury and recovery and use of EEG and MRI to study and evaluate prognostic factors for risk and recovery. It recognizes the work of others and discusses the UCDC's prior work and future research priorities.


Assuntos
Encéfalo , Eletroencefalografia , Hiperamonemia , Imageamento por Ressonância Magnética , Espectroscopia de Prótons por Ressonância Magnética , Distúrbios Congênitos do Ciclo da Ureia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/fisiopatologia , História do Século XXI , Hiperamonemia/diagnóstico por imagem , Hiperamonemia/história , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Distúrbios Congênitos do Ciclo da Ureia/diagnóstico por imagem , Distúrbios Congênitos do Ciclo da Ureia/história , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/fisiopatologia
4.
J Mother Child ; 24(2): 32-38, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33179600

RESUMO

Primary hyperammonaemia is a term to describe an elevation of ammonia in blood or plasma due to a defect within the urea cycle, which is the pathway responsible for ammonia detoxification and arginine biosynthesis. Urea cycle disorders (UCDs) are rare diseases caused by genetic defects affecting any of the six enzymes or two transporters that are directly involved in the urea cycle function.The clinical situation is variable and largely depends on the time of onset. Newborns who are often affected by hyper-ammonaemic encephalopathy carry a potential risk of severe brain damage, which may lead to death. Outside the neonatal period, symptoms are very unspecific but most often neurological (with wide variability), psychiatric and/or gastrointestinal. Early identification of patients is extremely important to start effective treatment modalities immediately. The acute management includes detoxification of ammonia, which often requires extracorporeal means such as haemodialysis, and the use of intravenous drugs that work as nitrogen scavengers. Long-term management of patients with UCDs consists of a low-protein diet, which needs to be balanced and supplemented to avoid deficiencies of essential amino acids, trace elements or vitamins and the use of nitrogen scavengers.The reader will find here a brief overview describing the most relevant aspects of the clinical management of UCDs in an attempt to raise awareness for this important group of rare diseases.


Assuntos
Amônia/sangue , Hiperamonemia/complicações , Hiperamonemia/diagnóstico , Hiperamonemia/fisiopatologia , Hiperamonemia/terapia , Diálise Renal/métodos , Distúrbios Congênitos do Ciclo da Ureia/sangue , Diagnóstico Precoce , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Resultado do Tratamento , Distúrbios Congênitos do Ciclo da Ureia/diagnóstico , Distúrbios Congênitos do Ciclo da Ureia/terapia
5.
Intern Med ; 59(20): 2465-2469, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33055469

RESUMO

Objective Rifaximin has become available for treating hyperammonemia in patients with hepatic encephalopathy. This study analyzed the changes in the body composition and nutritional status after long-term rifaximin therapy. Methods Twenty-one patients who underwent rifaximin therapy at 1,200 mg/day for more than 24 weeks were evaluated for the changes in the controlling nutritional status (CONUT) scores for the nutritional assessment, albumin-bilirubin (ALBI) scores for the liver function assessment, and skeletal muscle index (SMI) for the body composition assessment. Results There were 17 men and 4 women, with a mean age of 67.14±8.32 years. Eleven cases had a portosystemic shunt (52.3%), and 10 had hepatocellular carcinoma (47.6%). The Child-Pugh class was A in 9 cases (42.9%), B in 9 cases (42.9%), and C in 3 cases (14.2%). The blood ammonia levels in the rifaximin group improved significantly upon rifaximin therapy, from 124.76±28.68 µg/dL at baseline to 47.00±14.43 µg/dL after 2 weeks (p<0.001) and 49.81±15.02 µg/dL after 24 weeks (p<0.001). The CONUT scores improved significantly during rifaximin therapy, from 6.47±3.25 at baseline to 3.33±2.65 after 24 weeks (p=0.0007). The ALBI scores also improved significantly from -0.39±1.89 at baseline to -2.20±0.55 after 24 weeks (p=0.0002). The SMI scores showed that the body composition had been maintained in response to rifaximin therapy (50.20±7.67 at baseline and 51.29±7.62 after 24 weeks). Conclusion Rifaximin administration for hepatic encephalopathy improved the CONUT and ALBI scores. It may have a secondary effect on the improvement in the nutritional status and hepatic reserve.


Assuntos
Composição Corporal/efeitos dos fármacos , Encefalopatia Hepática/complicações , Hiperamonemia/tratamento farmacológico , Estado Nutricional/efeitos dos fármacos , Rifaximina/uso terapêutico , Idoso , Amônia/sangue , Bilirrubina/sangue , Carcinoma Hepatocelular/complicações , Feminino , Humanos , Hiperamonemia/etiologia , Hiperamonemia/fisiopatologia , Japão , Cirrose Hepática/complicações , Testes de Função Hepática , Neoplasias Hepáticas/complicações , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/fisiopatologia , Estudos Retrospectivos , Rifaximina/farmacologia , Albumina Sérica
6.
Clin Biochem ; 84: 63-72, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32569589

RESUMO

BACKGROUND: Ornithine transcarbamylase deficiency (OTCD) is pleomorphic congenital hyperammonemia, in which the prognosis of the patient is determined both by genotype and environmental factors. This study investigated the clinical and biochemical characteristics of OTCD patients with different prognosis. METHOD: Of 35 OTCD patients, six males deceased at the first disease-onset, 17 males survived and had controllable ammonia levels after treatment, and 12 females survived through the first disease-onset but had intractable hyperammonemia and high mortality. Fasting blood samples from patients collected at three disease stages were used for the analysis of amino acid (AA) profile, acylcarnitine profile, and micronutrients. Differences in profiles between patients and healthy controls and within patient groups were studied. RESULTS: All OTCD patients had accumulation of glutamine, homocitrulline, lysine, glutamate, cystathionine, and pipecolic acid, as well as deficiency of citrulline, tryptophan, threonine, and carnitine. For male non-survivors, most other AAs and long-chain acylcarnitines were elevated at disease onset, of which the levels of creatine, N-acetylaspartic acid, and homoarginine were remarkably high. Male survivors and female patients had most other AAs at low to normal levels. Compared with male survivors, female patients had much lower protein-intolerance, as indicated by significantly lower levels of protein consumption indicators, including essential AAs, 1-methylhistidine, acylcarnitines et al., but high levels of ammonia. Female patients still had significantly higher levels of citrulline, homocitrulline, and citrulline/arginine compared to male survivors. CONCLUSION: Unique profiles were observed in each group of OTCD patients, indicating specific physiological changes that happened to them.


Assuntos
Doença da Deficiência de Ornitina Carbomoiltransferase/metabolismo , Doença da Deficiência de Ornitina Carbomoiltransferase/fisiopatologia , Adolescente , Adulto , Amônia/sangue , Arginina/sangue , Criança , Pré-Escolar , China , Creatina/metabolismo , Feminino , Humanos , Hiperamonemia/fisiopatologia , Lisina/sangue , Masculino , Ornitina/uso terapêutico , Doença da Deficiência de Ornitina Carbomoiltransferase/sangue , Ureia/sangue , Adulto Jovem
7.
Crit Care Med ; 48(2): 218-224, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31939790

RESUMO

OBJECTIVES: Hyperammonemia is a key contributing factor for cerebral edema in acute liver failure. Continuous renal replacement therapy may help reduce ammonia levels. However, the optimal timing, mode, intensity, and duration of continuous renal replacement therapy in this setting are unknown. We aimed to study continuous renal replacement therapy use in acute liver failure patients and to assess its impact on hyperammonemia. DESIGN: Retrospective observational study. SETTING: ICU within a specialized liver transplant hospital. PATIENTS: Fifty-four patients with acute liver failure. INTERVENTIONS: Data were obtained from medical records and analyzed for patient characteristics, continuous renal replacement therapy use, ammonia dynamics, and outcomes. MAIN RESULTS: Forty-five patients (83%) had high grade encephalopathy. Median time to continuous renal replacement therapy commencement was 4 hours (interquartile range, 2-4.5) with 35 (78%) treated with continuous venovenous hemodiafiltration and 10 (22%) with continuous venovenous hemofiltration. Median hourly effluent flow rate was 43 mL/kg (interquartile range, 37-62). The median ammonia concentration decreased every day during treatment from 151 µmol/L (interquartile range, 110-204) to 107 µmol/L (interquartile range, 84-133) on day 2, 75 µmol/L (interquartile range, 63-95) on day 3, and 52 µmol/L (interquartile range, 42-70) (p < 0.0001) on day 5. The number of patients with an ammonia level greater than 150 µmol/L decreased on the same days from 26, to nine, then two, and finally none. Reductions in ammonia levels correlated best with the cumulative duration of therapy hours (p = 0.03), rather than hourly treatment intensity. CONCLUSIONS: Continuous renal replacement therapy is associated with reduced ammonia concentrations in acute liver failure patients. This effect is related to greater cumulative dose. These findings suggest that continuous renal replacement therapy initiated early and continued or longer may represent a useful approach to hyperammonemia control in acute liver failure patients.


Assuntos
Terapia de Substituição Renal Contínua/métodos , Hiperamonemia/etiologia , Hiperamonemia/terapia , Falência Hepática Aguda/complicações , APACHE , Adulto , Fatores Etários , Amônia/metabolismo , Peso Corporal , Feminino , Humanos , Hiperamonemia/fisiopatologia , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores Sexuais , Fatores de Tempo
8.
J Neurochem ; 154(1): 56-70, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31840253

RESUMO

Minimal hepatic encephalopathy (MHE) is a neuropsychiatric syndrome produced by central nervous system dysfunction subsequent to liver disease. Hyperammonemia and inflammation act synergistically to alter neurotransmission, leading to the cognitive and motor alterations in MHE, which are reproduced in rat models of chronic hyperammonemia. Patients with MHE show altered functional connectivity in different neural networks and a reduced response in the cognitive potential mismatch negativity (MMN), which correlates with attention deficits. The mechanisms by which MMN is altered in MHE remain unknown. The objectives of this work are as follows: To assess if rats with chronic hyperammonemia reproduce the reduced response in the MMN found in patients with MHE. Analyze the functional connectivity between the areas (CA1 area of the dorsal hippocampus, prelimbic cortex, primary auditory cortex, and central inferior colliculus) involved in the generation of the MMN and its possible alterations in hyperammonemia. Granger causality analysis has been applied to detect the net flow of information between the population neuronal activities recorded from a local field potential approach. Analyze if altered MMN response in hyperammonemia is associated with alterations in glutamatergic and GABAergic neurotransmission. Extracellular levels of the neurotransmitters and/or membrane expression of their receptors have been analyzed after the tissue isolation of the four target sites. The results show that rats with chronic hyperammonemia show reduced MMN response in hippocampus, mimicking the reduced MMN response of patients with MHE. This is associated with altered functional connectivity between the areas involved in the generation of the MMN. Hyperammonemia also alters membrane expression of glutamate and GABA receptors in hippocampus and reduces the changes in extracellular GABA and glutamate induced by the MMN paradigm of auditory stimulus in hippocampus of control rats. The changes in glutamatergic and GABAergic neurotransmission and in functional connectivity between the brain areas analyzed would contribute to the impairment of the MMN response in rats with hyperammonemia and, likely, also in patients with MHE.


Assuntos
Encéfalo/fisiopatologia , Potenciais Evocados Auditivos/fisiologia , Hiperamonemia/fisiopatologia , Vias Neurais/fisiopatologia , Transmissão Sináptica/fisiologia , Animais , Encefalopatia Hepática/fisiopatologia , Masculino , Ratos , Ratos Wistar
9.
Mol Genet Metab ; 129(3): 213-218, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31864849

RESUMO

Carnitine Uptake Defect (CUD) is an autosomal recessive disorder due to mutations in the SLC22A5 gene. Classically patients present in infancy with profound muscle weakness and cardiomyopathy with characteristic EKG findings. Later presentations include recurrent hypoketotic hypoglycemia, proximal limb girdle myopathy,and/or recurrent muscle pain. Newborn screening detects most of these clinical variants but in addition has identified maternal CUD often in asymptomatic women. We describe a family ascertained through 3 newborn screening (NBS) positive infants found to be unaffected themselves but in whom the mothers (sisters) were affected. There were also two affected children born to an affected male and his heterozygous wife who were false negatives on NBS but had increased fractional excretion of free carnitine in the urine. Analysis on a Next Generation Sequencing panel specifically designed to fully cover newborn screening disease targets showed a homozygous change in the five probands (SLC22A5; NM_003060:c.-149G > A; p.?). The mutation segregates with the CUD within the family. It is in the 5' UTR and has a frequency within the gnomAd database of 0.001198. Plasma carnitine was decreased and fractional excretion of free carnitine was increased in all affected individuals. Functional carnitine uptake studies in cultured skin fibroblasts of one proband showed carnitine uptake at the 5 µM concentration to be 6% of controls. Relative expression of OCTN2 mRNA to beta-actin mRNA by qRT-PCR was increased in a proband relative to controls by a factor of 465-fold. Western blotting revealed a 120 kDa protein band, as well as a weaker 240 kDa band in the proband, the significance of which is unknown at this time.


Assuntos
Regiões 5' não Traduzidas/genética , Cardiomiopatias/diagnóstico , Cardiomiopatias/genética , Carnitina/sangue , Carnitina/deficiência , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Doenças Musculares/diagnóstico , Doenças Musculares/genética , Membro 5 da Família 22 de Carreadores de Soluto/genética , Actinas/genética , Actinas/metabolismo , Transporte Biológico Ativo/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Carnitina/genética , Carnitina/metabolismo , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Lactente , Recém-Nascido , Masculino , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia , Mutação , Triagem Neonatal , Linhagem , Pele/citologia , Pele/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Sequenciamento do Exoma
10.
Orphanet J Rare Dis ; 14(1): 208, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31443672

RESUMO

BACKGROUND: Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is a rare disorder of urea cycle characterized by progressive pyramidal and cerebellar dysfunction, whose pathophysiology is not yet fully understood. Here we describe the spectrum of the long fibers involvement in HHH syndrome, attempting a correlation between clinical, electrophysiological and neuro-radiological data. METHODS: Nine HHH patients were longitudinally evaluated by clinical examination, neurophysiological assessment including motor (MEPs), somato-sensory evoked potentials (PESS) and nerve conduction velocity (NCV), brain and spinal cord MRI RESULTS: All patients had pyramidal dysfunction and 3/9 an overt spastic paraplegia. Mild to moderate cerebellar signs were found in 7/9, intellectual disability in 8/9. At lower limbs, MEPs resulted abnormal in 7/8 patients and PESS in 2/8; peripheral sensory-motor neuropathy was found in 1/9. MRI documented atrophic changes in supra-tentorial brain regions in 6/9 patients, cerebellum in 6/9, spinal cord in 3/7. CONCLUSIONS: A predominant corticospinal dysfunction is evident in HHH syndrome, along with milder cerebellar signs, intellectual disability of variable degree and rare peripheral neuropathy. Phenotypical similarities with other disorders affecting the urea cycle (argininemia and pyrroline-5-carboxylate synthetase deficiency) suggest possible common mechanisms contributing in the maintenance of the corticospinal tract integrity. HHH syndrome phenotype largely overlaps with complex Hereditary Spastic Paraplegias (HSPs), in the list of which it should be included, emphasizing the importance to screen all the unsolved cases of HSPs for metabolic biomarkers.


Assuntos
Hiperamonemia/metabolismo , Hiperamonemia/patologia , Ornitina/deficiência , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia , Adolescente , Adulto , Encéfalo/metabolismo , Encéfalo/fisiologia , Criança , Feminino , Humanos , Hiperamonemia/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação/genética , Condução Nervosa/fisiologia , Ornitina/metabolismo , Paraplegia Espástica Hereditária/metabolismo , Paraplegia Espástica Hereditária/patologia , Paraplegia Espástica Hereditária/fisiopatologia , Medula Espinal/metabolismo , Medula Espinal/fisiologia , Distúrbios Congênitos do Ciclo da Ureia/fisiopatologia , Adulto Jovem
11.
Am J Kidney Dis ; 74(3): 390-398, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31040091

RESUMO

Neurologic complications are common after solid-organ transplantation, occurring in one-third of patients. Immunosuppression-related neurotoxicity (involving calcineurin inhibitors and corticosteroids), opportunistic central nervous system infections, seizures, and delirium are some of the causes of neurologic symptoms following solid-organ transplantation. An uncommon often missed complication posttransplantation involves buildup of ammonia levels that can lead to rapid clinical deterioration even when treated. Ammonia levels are not routinely checked due to the myriad of other explanations for encephalopathy in a transplant recipient. A treatment of choice for severe hyperammonemia involves renal replacement therapy (RRT), but there are no guidelines on the mode or parameters of RRT for reducing ammonia levels. Hyperammonemia in a transplant recipient poses specific challenges beyond the actual condition because the treatment (RRT) involves significant hemodynamic fluctuations that may affect the graft. In this review, we describe a patient with posttransplantation hyperammonemia and discuss the pathways of ammonia metabolism, potential factors underlying the development of hyperammonemia posttransplantation, and choice of appropriate therapeutic options in these patients.


Assuntos
Hiperamonemia/fisiopatologia , Hiperamonemia/terapia , Transplante de Órgãos , Complicações Pós-Operatórias/fisiopatologia , Complicações Pós-Operatórias/terapia , Terapia de Substituição Renal , Humanos , Hiperamonemia/etiologia , Transplante de Órgãos/efeitos adversos , Complicações Pós-Operatórias/etiologia
12.
Brain Dev ; 41(7): 634-637, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30961960

RESUMO

Herpes simplex encephalitis (HSE) is a widely accepted risk factor for anti N-methyl-d-aspartate receptor (NMDAR) encephalitis. Association of inherited metabolic disease has never been reported in a patient with HSE and anti-NMDAR encephalitis. Herein, we report a case of pediatric HSE complicated by development of anti-NMDAR encephalitis; this patient showed subsequent recurrent, unexplained episodes of encephalopathy associated with hyperammonemia. The patient was diagnosed with lysinuric protein intolerance (LPI), a rare inborn metabolic disorder. Although it would be difficult to make conclusions regarding the casual link of HSE and anti-NMDAR encephalitis with LPI from a single case, there have been many reports that autoimmune diseases and immunologic abnormalities are frequently associated with LPI. Thus, we speculate that LPI may contribute to the development of anti-NMDAR encephalitis following HSE.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato/fisiopatologia , Hiperamonemia/fisiopatologia , Erros Inatos do Metabolismo dos Aminoácidos , Encefalite Antirreceptor de N-Metil-D-Aspartato/complicações , Encefalite Antirreceptor de N-Metil-D-Aspartato/metabolismo , Autoanticorpos , Encefalite por Herpes Simples/complicações , Feminino , Herpes Simples/complicações , Herpes Simples/fisiopatologia , Humanos , Hiperamonemia/complicações , Lactente , Receptores de N-Metil-D-Aspartato/imunologia
14.
Rev Gastroenterol Mex (Engl Ed) ; 84(2): 195-203, 2019.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-31014748

RESUMO

Understanding of the pathophysiology of hepatic encephalopathy has conditioned new treatment options. Ammonia detoxification in hepatic encephalopathy is regulated by two enzymes: glutaminase or glutamine synthetase. The first produces ammonia and the second detoxifies the ammonia, which is why treatments are aimed at glutaminase inhibition or glutamine synthetase activation. At present, we know that both enzymes are found not only in the liver, but also in the muscle, intestine, kidney, and brain. Therefore, current treatments can be directed at each enzyme at different sites. Awareness of those potential treatment sites makes different options of approach possible in the patient with hepatic encephalopathy, and each approach should be personalized.


Assuntos
Encefalopatia Hepática/fisiopatologia , Encefalopatia Hepática/terapia , Amônia/metabolismo , Ativadores de Enzimas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Glutamato-Amônia Ligase/efeitos dos fármacos , Glutaminase/antagonistas & inibidores , Humanos , Hiperamonemia/fisiopatologia , Hiperamonemia/terapia
15.
Clin Biochem ; 66: 100-102, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30753843

RESUMO

OBJECTIVES: Elevated levels of metabolites such as ammonia and propionylcarnitine in propionic acidemia (PA) lead to an increased reactive oxygen species (ROS) production which could activate and stabilize the epigenetic regulated hypoxia-inducible factor-1α (HIF-1α). In order to evaluate the DNA methylation status of the HIF-1α binding site in PA, we investigated the antioxidant gluthatione peroxidase 3 gene (GPX3) promoter region. DESIGN AND METHODS: Using leukocyte DNA extracted from bloodspots collected 2-4 days after birth from diet free newborns, the cytosine phosphodiester bond guanine (CpG) dinucleotides of a HIF-1α binding site (CGTTTTTTACG) in the promoter region of GPX3 was retrospectively analysed. Patients included 7 PA. and 7 healthy controls (KO) respectively. RESULTS: A demethylated TGTTTTTTATG allele was detected in 3 PA patients with blood ammonia (NH3) concentrations of 500, 595, and 987 umol/L respectively; a demethylated/partial methylated TGTTTTTTAC/TG allele in 4 PA patients (2 PA with blood NH3 = 213, 271 umol/L respectively); a partial methylated C/TGTTTTTTAC/TG allele in 5 healthy controls respectively; a partial methylated/methylated C/TGTTTTTTACG allele in 2 healthy controls. CONCLUSION: Our results suggest that at excess NH3, the DNA methylation status of the HIF-1α binding site of GPX3 in newborns with PA is demethylated (TGTTTTTTATG allele). However, the demethylated allele has to be confirmed as a statistically significant change in more patients.


Assuntos
Amônia/metabolismo , Metilação de DNA/fisiologia , Glutationa Peroxidase/genética , Hiperamonemia/fisiopatologia , Acidemia Propiônica/fisiopatologia , Sítios de Ligação/fisiologia , Desmetilação , Humanos , Recém-Nascido , Regiões Promotoras Genéticas/fisiologia
16.
Life Sci ; 218: 65-80, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30578865

RESUMO

Hepatic encephalopathy (HE) is a critical clinical complication. There is a consensus that ammonia plays a pivotal role in the pathogenesis of HE. Ammonia is a neurotoxin which induces a wide range of functional disturbances in the central nervous system (CNS). On the other hand, HE is associated with the increased free radical formation, tissue inflammation, disturbed neurotransmission, astrocytes swelling, brain edema, and brain herniation. In view of the severe CNS complications ensued HE, potential therapeutic points of intervention need to be vigorously investigated. A role for CNS mitochondrial damage and energy crisis has been considered in HE. It has been found that ammonia induces mitochondrial impairment as a result of a multifaceted interaction of different signaling molecules. Hence, ammonia-induced mitochondrial injury and compromised brain energy metabolism might play a vital role in the pathogenesis of ammonia neurotoxicity. This review focuses on the concept that mitochondrial dysfunction and cellular energy crisis indeed plays a critical role in the pathogenesis of hyperammonemia-induced brain injury. Further, it will highlight the potential therapeutic value of mitochondrial protecting agents and energy providers in the management of HE. The data collected in this review might provide clues to new therapeutic interventions aimed at minimizing HE-associated complications.


Assuntos
Lesões Encefálicas/complicações , Encéfalo/efeitos dos fármacos , Encefalopatia Hepática/prevenção & controle , Hiperamonemia/fisiopatologia , Mitocôndrias/efeitos dos fármacos , Taurina/farmacologia , Animais , Encéfalo/patologia , Metabolismo Energético , Encefalopatia Hepática/etiologia , Encefalopatia Hepática/patologia , Humanos , Mitocôndrias/patologia
18.
Mol Genet Metab ; 125(3): 235-240, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30197275

RESUMO

BACKGROUND: Continuous EEG studies demonstrate that neonates with seizures due to cerebral pathology, such as hypoxia ischemia, exhibit predominantly electrographic seizures (i.e. those only detected with EEG because they lack clinical features). Previous small case series demonstrate EEG changes and seizures during hyperammonemia associated with inborn errors of metabolism (IEM) but there are no reports utilizing continuous EEG in these conditions. OBJECTIVE: To characterize seizures and evaluate the utility of continuous EEG recording during hyperammonemia due to inborn errors of metabolism. METHODS: We retrospectively reviewed medical records and EEG tracings of neonates who presented with hyperammonemia due to inborn errors of metabolism who had continuous EEG and full medical records available for review, including follow up. RESULTS: Eight neonates with hyperammonemia were studied, 7 had urea cycle defects: Argininosuccinate lyase deficiency [3], (ornithine transcarbamylase deficiency [3], carbomyl phosphate synthase deficiency [1] and one had an organic acidemia: Methylmalonic acidemia [1]. Most common presentations were lethargy and poor feeding at 12-72 h of life. The highest blood ammonia level was 874 µmol/L (median); range 823-1647 µmol/L (normal value <50 µmol/L in term neonates). Seven were treated with hemodialysis in addition to nitrogen scavengers. Seven neonates had seizures; six had only electrographic seizures. Seizures initially occurred within 24-36 h of clinical presentation, sometimes with normal ammonia and glutamine levels. Neonates with seizures all lacked state changes on EEG. Inter burst interval duration correlated with degree of hyperammonemia. Two cases with normal plasma ammonia but increasing interburst interval duration were proven to have stroke by MRI. CONCLUSIONS: Seizures occur frequently in neonates with hyperammonemia; most can be detected only with continuous EEG. Seizures may occur when ammonia and glutamine levels are normal. Interburst interval duration is associated with ammonia levels or cerebral dysfunction from other brain pathology. Continuous EEG can be a useful tool for managing infants with hyperammonemia and may be essential for seizure management especially for infants in deep metabolic coma.


Assuntos
Amônia/sangue , Eletroencefalografia , Hiperamonemia/sangue , Erros Inatos do Metabolismo/sangue , Convulsões/sangue , Erros Inatos do Metabolismo dos Aminoácidos/sangue , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico por imagem , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Argininossuccinato Sintase/sangue , Acidúria Argininossuccínica/sangue , Acidúria Argininossuccínica/diagnóstico por imagem , Acidúria Argininossuccínica/fisiopatologia , Feminino , Glutamina/sangue , Humanos , Hiperamonemia/diagnóstico por imagem , Hiperamonemia/fisiopatologia , Hipóxia-Isquemia Encefálica/sangue , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Hipóxia-Isquemia Encefálica/fisiopatologia , Lactente , Recém-Nascido , Masculino , Erros Inatos do Metabolismo/diagnóstico por imagem , Erros Inatos do Metabolismo/fisiopatologia , Doença da Deficiência de Ornitina Carbomoiltransferase/sangue , Doença da Deficiência de Ornitina Carbomoiltransferase/diagnóstico por imagem , Doença da Deficiência de Ornitina Carbomoiltransferase/fisiopatologia , Convulsões/diagnóstico por imagem , Convulsões/fisiopatologia
19.
J Physiol Biochem ; 74(4): 523-530, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30058052

RESUMO

The aim was to determine the effects of enhanced availability of branched-chain amino acids (BCAAs; leucine, isoleucine, and valine) on ammonia detoxification to glutamine (GLN) and protein metabolism in two types of skeletal muscle under hyperammonemic conditions. Isolated soleus (SOL, slow-twitch) and extensor digitorum longus (EDL, fast-twitch) muscles from the left leg of white rats were incubated in a medium with 1 mM ammonia (NH3 group), BCAAs at four times the concentration of the controls (BCAA group) or high levels of both ammonia and BCAA (NH3 + BCAA group). The muscles from the right leg were incubated in basal medium and served as paired controls. L-[1-14C]leucine was used to estimate protein synthesis and leucine oxidation, and 3-methylhistidine release was used to evaluate myofibrillar protein breakdown. We observed decreased protein synthesis and glutamate and α-ketoglutarate (α-KG) levels and increased leucine oxidation, GLN levels, and GLN release into medium in muscles in NH3 group. Increased leucine oxidation, release of branched-chain keto acids and GLN into incubation medium, and protein synthesis in EDL were observed in muscles in the BCAA group. The addition of BCAAs to medium eliminated the adverse effects of ammonia on protein synthesis and adjusted the decrease in α-KG found in the NH3 group. We conclude that (i) high levels of ammonia impair protein synthesis, activate BCAA catabolism, enhance GLN synthesis, and decrease glutamate and α-KG levels and (ii) increased BCAA availability enhances GLN release from muscles and attenuates the adverse effects of ammonia on protein synthesis and decrease in α-KG.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Hiperamonemia/metabolismo , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Amônia/intoxicação , Animais , Radioisótopos de Carbono , Ciclo do Ácido Cítrico/efeitos dos fármacos , Glutamina/agonistas , Glutamina/metabolismo , Hiperamonemia/enzimologia , Hiperamonemia/fisiopatologia , Técnicas In Vitro , Ácidos Cetoglutáricos/metabolismo , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Metilistidinas/metabolismo , Fibras Musculares de Contração Rápida/efeitos dos fármacos , Fibras Musculares de Contração Rápida/enzimologia , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Fibras Musculares de Contração Lenta/enzimologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Especificidade de Órgãos , Concentração Osmolar , Oxirredução , Biossíntese de Proteínas/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ratos Wistar
20.
Metab Brain Dis ; 33(4): 1335-1342, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29725955

RESUMO

Hyperammonemia in adults is generally associated with cerebral edema, decreased cerebral metabolism, and increased cerebral blood flow. The aim of this study was to evaluate the association between non-hepatic hyperammonemia and intracranial hypertension assessed by Doppler flowmetry and measurement of the optic nerve sheath. A prospective cohort study in critically ill patients hospitalized in intensive care units of a University Hospital between March 2015 and February 2016. Clinical data and severity scores were collected and the Glasgow coma scale was recorded. Serial serum ammonia dosages were performed in all study patients. Transcranial Doppler evaluation was carried out for the first 50 consecutive results of each stratum of ammonemia: normal (<35 µmol/L), mild hyperammonemia (≥35 µmol/L and < 50 µmol/L), moderate hyperammonemia (≥50 µmol/L and < 100 µmol/L), and severe hyperammonemia (≥100 µmol/L). The measurement of the optic nerve sheath was performed at the same time as the Doppler examination if the patient scored less than 8 on the Glasgow coma scale. There was no difference in flow velocity in the cerebral arteries between patients with and without hyperammonemia. Patients with hyperammonemia presented longer ICU stay. Optic nerve sheath thickness was higher in the group with severe hyperammonemia and this group presented an association with intracranial hypertension. Higher mortality was observed in the severe hyperammonemia group. There was an association between severe hyperammonemia and signs of intracranial hypertension. No correlation was found between ammonia levels and cerebral blood flow velocity through the Doppler examination.


Assuntos
Circulação Cerebrovascular/fisiologia , Hiperamonemia/diagnóstico por imagem , Hipertensão Intracraniana/diagnóstico por imagem , Nervo Óptico/diagnóstico por imagem , Adulto , Idoso , Estado Terminal , Feminino , Escala de Coma de Glasgow , Humanos , Hiperamonemia/fisiopatologia , Hipertensão Intracraniana/fisiopatologia , Pressão Intracraniana/fisiologia , Masculino , Pessoa de Meia-Idade , Nervo Óptico/fisiopatologia , Estudos Prospectivos , Índice de Gravidade de Doença , Ultrassonografia Doppler Transcraniana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...