Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Inherit Metab Dis ; 47(1): 50-62, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37026568

ABSTRACT

Urea cycle defects (UCDs) are severe inherited metabolic diseases with high unmet needs which present a permanent risk of hyperammonaemic decompensation and subsequent acute death or neurological sequelae, when treated with conventional dietetic and medical therapies. Liver transplantation is currently the only curative option, but has the potential to be supplanted by highly effective gene therapy interventions without the attendant need for life-long immunosuppression or limitations imposed by donor liver supply. Over the last three decades, pioneering genetic technologies have been explored to circumvent the consequences of UCDs, improve quality of life and long-term outcomes: adenoviral vectors, adeno-associated viral vectors, gene editing, genome integration and non-viral technology with messenger RNA. In this review, we present a summarised view of this historical path, which includes some seminal milestones of the gene therapy's epic. We provide an update about the state of the art of gene therapy technologies for UCDs and the current advantages and pitfalls driving future directions for research and development.


Subject(s)
Liver Transplantation , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Humans , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Quality of Life , Urea/metabolism , Living Donors , Genetic Therapy , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/therapy , Urea Cycle Disorders, Inborn/complications
2.
Curr Opin Clin Nutr Metab Care ; 27(2): 184-191, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37938118

ABSTRACT

PURPOSE OF REVIEW: Urea cycle disorders (UCDs) cause elevations in ammonia which, when severe, cause irreversible neurologic injury. Most patients with UCDs are diagnosed as neonates, though mild UCDs can present later - even into adulthood - during windows of high physiologic stress, like critical illness. It is crucial for clinicians to understand when to screen for UCDs and appreciate how to manage these disorders in order to prevent devastating neurologic injury or death. RECENT FINDINGS: Hyperammonemia, particularly if severe, causes time- and concentration-dependent neurologic injury. Mild UCDs presenting in adulthood are increasingly recognized, so broader screening in adults is recommended. For patients with UCDs, a comprehensive, multitiered approach to management is needed to prevent progression and irreversible injury. Earlier exogenous clearance is increasingly recognized as an important complement to other therapies. SUMMARY: UCDs alter the core pathway for ammonia metabolism. Screening for mild UCDs in adults with unexplained neurologic symptoms can direct care and prevent deterioration. Management of UCDs emphasizes decreasing ongoing ammonia production, avoiding catabolism, and supporting endogenous and exogenous ammonia clearance. Core neuroprotective and supportive critical care supplements this focused therapy.


Subject(s)
Hyperammonemia , Urea Cycle Disorders, Inborn , Adult , Humans , Ammonia , Critical Illness , Hyperammonemia/etiology , Longitudinal Studies , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/therapy
3.
Arch. argent. pediatr ; 121(3): e202202614, jun. 2023. tab
Article in English, Spanish | LILACS, BINACIS | ID: biblio-1435904

ABSTRACT

La hiperamonemia constituye una emergencia médica. No existen publicaciones que hagan referencia a la disponibilidad de recursos, insumos y conocimientos necesarios para el manejo inicial de esta por parte del pediatra en nuestro país, pero, según la experiencia de los autores, los recursos necesarios no se encuentran disponibles los 365 días del año en una gran porción de nuestro territorio. Sobre la base de este estado de situación, de una revisión bibliográfica internacional sobre el tema y de la experiencia de los autores, se elaboraron una serie de recomendaciones para el manejo pediátrico inicial de esta emergencia, que tienen como objetivo poder reducir las deficiencias, permitir una sospecha clínica adecuada que lleve a un diagnóstico y tratamiento de emergencia oportunos, con utilización racional de recursos farmacológicos (algunos de ellos de alto costo), para reducir la morbimortalidad que asocia la patología.


Hyperammonemia is a medical emergency. There are no publications regarding the availability of resources, supplies, and knowledge necessary for the initial management of hyperammonemia by pediatricians in Argentina; however, according to the authors' experience, the necessary resources are not available all year round in a large portion of our territory. Based on such state of affairs, an international bibliographic review on this topic and the authors' experience, we developed a series of recommendations for the initial pediatric management of this emergency, with the objective of reducing deficiencies, allowing adequate clinical suspicion leading to a timely diagnosis and emergency management and a rational use of pharmacological resources (some of which are costly) to reduce the morbidity and mortality associated with hyperammonemia.


Subject(s)
Humans , Infant , Child, Preschool , Child , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Argentina
4.
Bone ; 169: 116684, 2023 04.
Article in English | MEDLINE | ID: mdl-36709914

ABSTRACT

Carbonic anhydrase II deficiency (OMIM # 259730), initially called "osteopetrosis with renal tubular acidosis and cerebral calcification syndrome", reveals an important role for the enzyme carbonic anhydrase II (CA II) in osteoclast and renal tubule function. Discovered in 1972 and subsequently given various names, CA II deficiency now describes >100 affected individuals encountered predominantly from the Middle East and Mediterranean region. In 1983, CA II deficiency emerged as the first osteopetrosis (OPT) understood metabolically, and in 1991 the first understood molecularly. CA II deficiency is the paradigm OPT featuring failure of osteoclasts to resorb bone due to inability to acidify their pericellular milieu. The disorder presents late in infancy or early in childhood with fracturing, developmental delay, weakness, short stature, and/or cranial nerve compression and palsy. Mental retardation is common. The skeletal findings may improve by adult life, and CA II deficiency can be associated with a normal life-span. Therefore, it has been considered an "intermediate" type of OPT. In CA II deficiency, OPT is uniquely accompanied by renal tubular acidosis (RTA) of proximal, distal, or combined type featuring hyperchloremic metabolic acidosis, rarely with hypokalemia and paralysis. Cerebral calcification uniquely appears in early childhood. The etiology is bi-allelic loss-of-function mutations of CA2 that encodes CA II. Prenatal diagnosis requires mutational analysis of CA2. Although this enzymopathy reveals how CA II is important for the skeleton and kidney tubule, the pathogenesis of the mental subnormality and cerebral calcification is less well understood. Several mouse models of CA II deficiency have shown growth hormone deficiency, yet currently there is no standard pharmacologic therapy for patients. Treatment of the systemic acidosis is often begun when growth is complete. Although CA II deficiency is an "osteoclast-rich" OPT, and therefore transplantation of healthy osteoclasts can improve the skeletal disease, the RTA and central nervous system difficulties persist.


Subject(s)
Acidosis, Renal Tubular , Calcinosis , Carbonic Anhydrase II , Carbonic Anhydrases , Intellectual Disability , Osteopetrosis , Urea Cycle Disorders, Inborn , Animals , Child, Preschool , Female , Humans , Mice , Pregnancy , Acidosis, Renal Tubular/complications , Acidosis, Renal Tubular/genetics , Calcinosis/genetics , Carbonic Anhydrases/genetics , Intellectual Disability/genetics , Osteopetrosis/genetics , Urea Cycle Disorders, Inborn/complications , Carbonic Anhydrase II/deficiency
5.
Arch Argent Pediatr ; 121(3): e202202614, 2023 06 01.
Article in English, Spanish | MEDLINE | ID: mdl-36287611

ABSTRACT

Hyperammonemia is a medical emergency. There are no publications regarding the availability of resources, supplies, and knowledge necessary for the initial management of hyperammonemia by pediatricians in Argentina; however, according to the authors' experience, the necessary resources are not available all year round in a large portion of our territory. Based on such state of affairs, an international bibliographic review on this topic and the authors' experience, we developed a series of ecommendations for the initial pediatric management of this emergency, with the objective of reducing deficiencies, allowing adequate clinical suspicion leading to a timely diagnosis and emergency management and a rational use of pharmacological resources (some of which are costly) to reduce the morbidity and mortality associated with hyperammonemia.


La hiperamonemia constituye una emergencia médica. No existen publicaciones que hagan referencia a la disponibilidad de recursos, insumos y conocimientos necesarios para el manejo inicial de esta por parte del pediatra en nuestro país, pero, según la experiencia de los autores, los recursos necesarios no se encuentran disponibles los 365 días del año en una gran porción de nuestro territorio. Sobre la base de este estado de situación, de una revisión bibliográfica internacional sobre el tema y de la experiencia de los autores, se elaboraron una serie de recomendaciones para el manejo pediátrico inicial de esta emergencia, que tienen como objetivo poder reducir las deficiencias, permitir una sospecha clínica adecuada que lleve a un diagnóstico y tratamiento de emergencia oportunos, con utilización racional de recursos farmacológicos (algunos de ellos de alto costo), para reducir la morbimortalidad que asocia la patología.


Subject(s)
Hyperammonemia , Urea Cycle Disorders, Inborn , Humans , Child , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Argentina
6.
J Inherit Metab Dis ; 46(2): 220-231, 2023 03.
Article in English | MEDLINE | ID: mdl-36266255

ABSTRACT

The SARS-CoV-2 pandemic challenges healthcare systems worldwide. Within inherited metabolic disorders (IMDs) the vulnerable subgroup of intoxication-type IMDs such as organic acidurias (OA) and urea cycle disorders (UCD) show risk for infection-induced morbidity and mortality. This study (observation period February 2020 to December 2021) evaluates impact on medical health care as well as disease course and outcome of SARS-CoV-2 infections in patients with intoxication-type IMDs managed by participants of the European Registry and Network for intoxication type metabolic diseases Consortium (E-IMD). Survey's respondents managing 792 patients (n = 479 pediatric; n = 313 adult) with intoxication-type IMDs (n = 454 OA; n = 338 UCD) in 14 countries reported on 59 (OA: n = 36; UCD: n = 23), SARS-CoV-2 infections (7.4%). Medical services were increasingly requested (95%), mostly alleviated by remote technologies (86%). Problems with medical supply were scarce (5%). Regular follow-up visits were reduced in 41% (range 10%-50%). Most infected individuals (49/59; 83%) showed mild clinical symptoms, while 10 patients (17%; n = 6 OA including four transplanted MMA patients; n = 4 UCD) were hospitalized (metabolic decompensation in 30%). ICU treatment was not reported. Hospitalization rate did not differ for diagnosis or age group (p = 0.778). Survival rate was 100%. Full recovery was reported for 100% in outpatient care and 90% of hospitalized individuals. SARS-CoV-2 impacts health care of individuals with intoxication-type IMDs worldwide. Most infected individuals, however, showed mild symptoms and did not require hospitalization. SARS-CoV-2-induced metabolic decompensations were usually mild without increased risk for ICU treatment. Overall prognosis of infected individuals is very promising and IMD-specific or COVID-19-related complications have not been observed.


Subject(s)
COVID-19 , Metabolic Diseases , Urea Cycle Disorders, Inborn , Adult , Humans , Child , SARS-CoV-2 , Pandemics , Urea Cycle Disorders, Inborn/complications
7.
Int J Dev Neurosci ; 82(8): 772-788, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36129623

ABSTRACT

Urea cycle disorders (UCD) are a group of genetic diseases caused by deficiencies in the enzymes and transporters involved in the urea cycle. The impairment of the cycle results in ammonia accumulation, leading to neurological dysfunctions and poor outcomes to affected patients. The aim of this study is to investigate and describe UCD patients' principal clinical and biochemical presentations to support professionals on urgent diagnosis and quick management, aiming better outcomes for patients. We explored medical records of 30 patients diagnosed in a referral center from Brazil to delineate UCD clinical and biochemical profile. Patients demonstrated a range of signs and symptoms, such as altered levels of consciousness, acute encephalopathy, seizures, progressive loss of appetite, vomiting, coma, and respiratory distress, in most cases combined with high levels of ammonia, which is an immediate biomarker, leading to a UCD suspicion. The most prevalent UCD detected were ornithine transcarbamylase deficiency, followed by citrullinemia type 1, hyperargininemia, carbamoyl phosphate synthase 1 deficiency, and argininosuccinic aciduria. Clinical symptoms were highly severe, being the majority developmental and neurological disabilities, with 20% of death rate. Laboratory analysis revealed high levels of ammonia (mean ± SD: 860 ± 470 µmol/L; reference value: ≤80 µmol/L), hypoglycemia, metabolic acidosis, and high excretion of orotic acid in the urine (except in carbamoyl phosphate synthetase 1 [CPS1] deficiency). We emphasize the need of urgent identification of UCD clinical and biochemical conditions, and immediate measurement of ammonia, to enable the correct diagnosis and increase the chances of patients' survival, minimizing neurological and psychomotor damage caused by hepatic encephalopathy.


Subject(s)
Hepatic Encephalopathy , Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hepatic Encephalopathy/complications , Hepatic Encephalopathy/diagnosis , Ammonia , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase Deficiency Disease/genetics
8.
Front Immunol ; 13: 861516, 2022.
Article in English | MEDLINE | ID: mdl-35711415

ABSTRACT

The hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is a rare autosomal recessive inborn error of the urea cycle caused by mutations in the SLC25A15 gene. Besides the well-known metabolic complications, patients often present intercurrent infections associated with acute hyperammonemia and metabolic decompensation. However, it is currently unknown whether intercurrent infections are associated with immunological alterations besides the known metabolic imbalances. Herein, we describe the case of a 3-years-old girl affected by the HHH syndrome caused by two novel SLC25A15 gene mutations associated with immune phenotypic and functional alterations. She was admitted to the hospital with an episode of recurrent otitis, somnolence, confusion, and lethargy. Laboratory tests revealed severe hyperammonemia, elevated serum levels of liver transaminases, hemostasis alterations, hyperglutaminemia and strikingly increased orotic aciduria. Noteworthy, serum protein electrophoresis showed a reduction in the gamma globulin fraction. Direct sequencing of the SLC25A15 gene revealed two heterozygous non-conservative substitutions in the exon 5: c.649G>A (p.Gly217Arg) and c.706A>G (p.Arg236Gly). In silico analysis indicated that both mutations significantly impair protein structure and function and are consistent with the patient clinical status confirming the diagnosis of HHH syndrome. In addition, the immune analysis revealed reduced levels of serum IgG and striking phenotypic and functional alterations in the T and B cell immune compartments. Our study has identified two non-previously described mutations in the SLC25A15 gene underlying the HHH syndrome. Moreover, we are reporting for the first time functional and phenotypic immunologic alterations in this rare inborn error of metabolism that would render the patient immunocompromised and might be related to the high frequency of intercurrent infections observed in patients bearing urea cycle disorders. Our results point out the importance of a comprehensive analysis to gain further insights into the underlying pathophysiology of the disease that would allow better patient care and quality of life.


Subject(s)
Hyperammonemia , Urea Cycle Disorders, Inborn , Amino Acid Transport Systems, Basic/genetics , Child, Preschool , Female , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Mitochondrial Membrane Transport Proteins , Ornithine/deficiency , Quality of Life , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
9.
J Med Case Rep ; 16(1): 96, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35249549

ABSTRACT

BACKGROUND: Hyperammonemia is a medical condition described as increased or elevated serum ammonia levels. High serum levels of ammonia can cause neurotoxicity. Sudden onset severe hyperammonemia may cause severe encephalopathy with brain damage. It can result in cerebral edema, emesis, seizures, hypotonia, and death. We report a young postpartum woman who had a sudden rise in serum ammonia levels after vaginal delivery. CASE PRESENTATION: A 24-year-old, married, postpartum Pakistani woman was admitted to the intensive care unit through the emergency department, with complaints of fever, severe abdominal pain with distension, and altered levels of consciousness. The patient had a medical history of spontaneous vaginal delivery 2 weeks before this hospital admission, after which she gradually developed the above symptoms. However, the patient's past medical history was unremarkable with no hepatic disease, but her investigations revealed a progressive rise in serum ammonia levels. In the intensive care unit, she developed generalized tonic-clonic seizures. This was followed by a coma, tonsillar herniation, and death. CONCLUSION: Postpartum hyperammonemia is a rare entity. It is a critical illness and must be evaluated for underlying metabolic disorders. Early diagnosis and treatment may result in better outcomes and reduced mortality among postpartum women with hyperammonemia.


Subject(s)
Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Adult , Coma/etiology , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/etiology , Hyperammonemia/therapy , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Postpartum Period , Urea Cycle Disorders, Inborn/complications , Young Adult
10.
Mol Genet Metab ; 135(1): 3-14, 2022 01.
Article in English | MEDLINE | ID: mdl-34996714

ABSTRACT

Inborn errors of metabolism causing stroke (ischemic or haemorrhagic) or stroke-like episodes (e.g., that are also called "metabolic strokes" and include acute brain lesions not related with alterations of blood flow) cover a wide range of diseases in which acute metabolic decompensations after trigger events (e.g., fever, dehydration, sepsis etc.) may have a variable frequency. The early diagnosis of these conditions is essential because, despite their rarity, effective symptomatic treatments may be available for acute settings (e.g., arginine for Mitochondrial myopathy, Encephalopathy, Lactic Acidosis, and Stroke-like episodes- MELAS) while in other cases disease modifying therapies may be useful to prevent stroke occurrence, recurrence, or relapse (e.g., Fabry disease). The detection of a non-vascular distribution of lesions and the diffuse use of 1HMRS are often diriment in the differential of ischemic and metabolic strokes. This review summarized the main clinical features and the pathophysiological mechanisms of stroke and stroke-like episodes in inborn errors of metabolism presenting with stroke as part of natural history of the disease. These conditions belong to different etiological groups, such as organic acidurias, mitochondrial encephalopathies, homocystinuria and remethylation disorders, urea cycle disorders, lysosomal diseases (e.g. Fabry disease, glycogen storage disease), congenital disorders of glycosylation, neurotransmitter disorders, adenosine deaminase 2 deficiency and few other neurometabolic disorders.


Subject(s)
Fabry Disease , MELAS Syndrome , Mitochondrial Myopathies , Stroke , Urea Cycle Disorders, Inborn , Fabry Disease/complications , Humans , MELAS Syndrome/genetics , Stroke/etiology , Stroke/metabolism , Urea Cycle Disorders, Inborn/complications
11.
Intern Med ; 61(9): 1387-1392, 2022 May 01.
Article in English | MEDLINE | ID: mdl-34670888

ABSTRACT

A 36-year-old man experienced severely impaired consciousness twice after drinking because of hyperammonemia. No abnormal blood tests were found other than ammonia levels. However, magnetic resonance imaging (MRI) showed atrophy of the brain parenchyma. One the second occasion, the patient suffered severe impairment of consciousness, and because of seizures and glossoptosis, mechanical ventilation was started. Urea cycle disorders (UCDs) were assumed to be involved. Genetic testing revealed a monoallelic mutation of the carbamoyl phosphate synthase 1 (CPS1) gene. When transient hyperammonemia of unknown cause occurs repeatedly in adults, an active investigation for UCDs should be conducted.


Subject(s)
Carbamoyl-Phosphate Synthase I Deficiency Disease , Hyperammonemia , Urea Cycle Disorders, Inborn , Adult , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/complications , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamyl Phosphate , Consciousness , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Male , Mutation/genetics , Urea Cycle Disorders, Inborn/complications
12.
Intern Med ; 61(4): 553-557, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34433721

ABSTRACT

A 49-year-old Japanese man had shown developmental delay, learning difficulties, epilepsy, and slowly progressive gait disturbance in elementary school. At 46 years old, he experienced repeated drowsiness with or without generalized convulsions, and hyperammonemia was detected. Brain magnetic resonance imaging detected multiple cerebral white matter lesions. An electroencephalogram showed diffuse slow basic activities with 2- to 3-Hz δ waves. Genetic tests confirmed a diagnosis of hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome. Leukoencephalopathy was resolved following the administration of L-arginine and lactulose with a decrease in plasma ammonia levels and glutamine-glutamate peak on magnetic resonance spectroscopy. Leukoencephalopathy in HHH syndrome may be reversible with the resolution of hyperammonemia-induced glutamine toxicity.


Subject(s)
Hyperammonemia , Leukoencephalopathies , Urea Cycle Disorders, Inborn , Ammonia , Child , Humans , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Male , Middle Aged , Ornithine/deficiency , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
13.
BMJ Case Rep ; 14(7)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34210698

ABSTRACT

Hyperornithinaemia-hyperammonaemia-homocitrullinuria (HHH) syndrome is a rare inherited metabolic disorder of the urea cycle. Few reports exist to guide practices during pregnancy and fetal delivery. Yet, with affected patients often surviving into reproductive age, appropriate management of the peripartum phase is essential to ensure positive maternal and fetal outcomes.Reassuringly, the vast majority of offspring of parturients with HHH syndrome have normal developmental outcomes; yet as seen here, fetal growth restriction does appear more frequently. Furthermore, in addition to the absent fetal corpus callosum observed in this case, other fetal cerebral abnormalities, including speech delay and intellectual impairment, have been recognised.Unregulated dietary intake is one proposed factor for the observed disruption in fetal growth and early cerebral development. These stipulations not only reinforce the importance of extensive planning and teamwork, but also demonstrate the importance of timely intervention by a metabolic dietician and dietary compliance in the early organogenesis stage of pregnancy.


Subject(s)
Hyperammonemia , Urea Cycle Disorders, Inborn , Citrulline , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Ornithine/deficiency , Pregnancy , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
14.
Liver Transpl ; 27(12): 1799-1810, 2021 12.
Article in English | MEDLINE | ID: mdl-34058057

ABSTRACT

Liver transplantation (LT) for children with urea cycle disorders (UCDs) is capable of correcting the enzymatic defect and preventing progressive neurologic injury. We describe the characteristics and outcomes of pediatric LT recipients with UCDs. We identified all pediatric (<18 years) LT candidates with UCDs in the United Network for Organ Sharing (UNOS) database (February 2002 to September 2020). Multivariable Cox and logistic regression were used to determine risk factors for graft loss and cognitive delay, respectively. Of 424 patients, 1.9% (8/424) experienced waitlist mortality and 95.0% underwent LT (403/424). The most frequently encountered UCDs in our cohort were ornithine transcarbamylase deficiency (46.2%), citrullinemia (20.3%), and argininosuccinic aciduria (ASA; 12.9%). The 1-, 3-, and 5-year graft survival rates were 90.4%, 86.3%, and 85.2%, respectively. Multivariable analysis showed a decreased risk of graft loss with increasing weight at LT (adjusted hazard ratio [aHR], 0.96; 95% confidence interval [CI], 0.94-0.99; P = 0.02), male sex (aHR, 0.49; 95% CI, 0.28-0.85; P = 0.01), and ASA diagnosis (aHR, 0.29; 95% CI, 0.09-0.98; P = 0.047), when adjusting for location (intensive care/hospital/home) and graft type (both P ≥ 0.65). In multivariable logistic regression, waitlist time (adjusted odds ratio [aOR], 1.10; 95% CI, 1.02-1.17; P = 0.009) and male sex (aOR, 1.71; 95% CI, 1.02-2.88; P = 0.04) were associated with increased odds of long-term cognitive delay. Waitlist duration is associated with a long-term risk of cognitive delay. Given excellent long-term outcomes, early LT evaluation should be considered in all children with UCDs to prevent progressive neurologic injury and optimize cognitive outcomes.


Subject(s)
Liver Transplantation , Urea Cycle Disorders, Inborn , Child , Graft Survival , Humans , Liver Transplantation/adverse effects , Male , Risk Factors , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/epidemiology , Waiting Lists
15.
Galicia clin ; 82(Supl. 1): s23-s25, Febrero 2021.
Article in Spanish | IBECS | ID: ibc-220933

ABSTRACT

La hiperamonemia es una de las complicaciones agudas más graves que se puede presentar en diversos trastornos congénitos del metabolismo como los defectos en el ciclo de la urea, las acidemias orgánicas o en las alteraciones en la oxidación de los ácidos grasos. Su reconocimiento y tratamiento es una emergencia clínica, dado que el efecto neurotóxico es directamente proporcional al tiempo de exposición a niveles elevados de amonio. Presentamos el caso clínico de un paciente de 30 años con deterioro del nivel de consciencia y elevación del amonio plasmático de 24 horas de evolución. (AU)


Hyperammonemia is one of the most severe acute complication that can occur in various congenital metabolic conditions such as the urea cycle disorders, organic acidemias or fatty acids oxidation defects. The prompt recognition and treatment is a clinical emergency, since the neurotoxic effect is directly proportional to the time of exposure to high levels of ammonia. We report the clinical case of a 30-year-old patient with a decreased level of consciousness and a 24-hour evolution of high plasma ammonia. (AU)


Subject(s)
Humans , Male , Adult , Hyperammonemia , Fatty Acids , Hypoglycemia , Urea Cycle Disorders, Inborn/complications , Oxidation , Uric Acid
16.
J Neuroimaging ; 31(1): 144-150, 2021 01.
Article in English | MEDLINE | ID: mdl-32920938

ABSTRACT

BACKGROUND AND PURPOSE: This study aimed to assess characteristic regions of MRI involvement utilizing diffusion weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) at urea cycle disorder (UCD) diagnosis to determine the possible association between initial MRI patterns within 10 days of the first hyperammonemia episode, serum ammonia levels, and severity of neurological outcome based on clinical follow-up of >30 days. METHODS: Ten patients with UCDs (4 females; median age: 5.4 years, age range: 6 days-54 years) were included who underwent MRI during a first episode of hyperammonemia. The topographical distribution of the DWI and FLAIR abnormalities in the cerebral cortex, deep gray matter, white matter, posterior limb of internal capsule, cerebral peduncle, and cerebellum was evaluated. Possible correlations between the brain injury patterns on DWI/FLAIR images, serum ammonia levels, and severity of neurological outcome were investigated by a trend correlation. RESULTS: The UCD cohort (n = 10) involved four ornithine transcarbamoylase deficiencies, four argininosuccinic aciduria, one carbomoylphosphate synthetase deficiency, and one citrullinemia type-1. The observed trend in the distribution of DWI abnormalities as the severity of neurological sequela outcome increased was with diffuse cerebral cortex or corpus striatum involvement. Patients with initial peak serum ammonia ≥450 µmol/L had a grade 2 to 4 outcome, and those with peak ammonia <450 µmol/L had a grade 0 or 1 outcome. CONCLUSIONS: The presence of more severe neurological outcome could be associated with diffuse cerebral cortex or corpus striatum involvement on DWI and high serum ammonia levels in patients with UCD.


Subject(s)
Diffusion Magnetic Resonance Imaging , Neuroimaging , Urea Cycle Disorders, Inborn/diagnostic imaging , Adult , Child , Child, Preschool , Disease Progression , Female , Humans , Male , Middle Aged , Retrospective Studies , Urea Cycle Disorders, Inborn/complications , Young Adult
17.
Biomed Res Int ; 2020: 5690915, 2020.
Article in English | MEDLINE | ID: mdl-32934962

ABSTRACT

During Jan. 2016-Dec. 2019, nine Chinese patients from eight unrelated families were diagnosed with neonatal-onset UCDs by targeted panel sequencing or whole-exome sequencing (WES). Their clinical manifestations, biochemical features, 180-day-age outcomes, and molecular genetic characteristics were reviewed retrospectively. NGS-based tests revealed 7 patients diagnosed with ornithine transcarbamylase deficiency (OTCD) and 2 with carbamoylphosphate synthetase I deficiency (CPS1D). The spectrum of the clinical presentation of nine affected individuals progressed from unspecific symptoms like poor feeding to somnolence, coma, and death. All patients presented with an acute hyperammonemia. The most robust metabolic pattern in OTCD was hyperglutaminemic hyperammonemia with high concentration of urine orotic acid, and it was reported in six patients. Of ten variants found on the OTC gene and CPS1 gene, 3 were novel: (c.176T>C (p.L59P)) in the OTC gene, c.2938G>A (p.G980S) and c.3734T>A (p.L1245H) in the CPS1 gene. There was a high mortality rate of 77.78% (7/9) for all the defects combined. An OTC-deficient male and a CPS1-deficient female survived from episodes of hyperammonemia. Although prompt recognition of UCD and the use of alternative pathway therapy in addition to provision of appropriate nutrition and dialysis improved survival, the overall outcomes for the neonatal-onset type are poor in China.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Hyperammonemia/genetics , Ornithine Carbamoyltransferase/genetics , Urea Cycle Disorders, Inborn/genetics , Age of Onset , China , Female , High-Throughput Nucleotide Sequencing , Humans , Hyperammonemia/complications , Hyperammonemia/metabolism , Hyperammonemia/pathology , Infant, Newborn , Male , Metabolomics/methods , Mutation/genetics , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Exome Sequencing
18.
Paediatr Anaesth ; 30(7): 780-791, 2020 07.
Article in English | MEDLINE | ID: mdl-32375202

ABSTRACT

BACKGROUND: Urea cycle disorders are congenital metabolism errors that affect ammonia elimination. Clinical signs and prognosis are strongly influenced by peak ammonia levels. Numerous triggers associated with metabolic decompensation have been described with many of them, including fasting or stress, being related to the perioperative period. AIMS: We aimed to assess perioperative complications in pediatric patients with urea cycle disorders requiring general anesthesia in our center. METHODS: We reviewed the clinical history of all the pediatric patients with a confirmed urea cycle disorders diagnosis requiring surgery or a diagnostic procedure with anesthesia between January 2002 and June 2018. RESULTS: We included 33 operations (major surgery, minor surgery, and diagnostic procedures) carried out on 10 patients via different anesthetic techniques. We observed the following complications: intraoperative hyperglycemia in one case, postoperative vomiting in eight cases, and slightly increased postoperative ammonia levels (54, 59, and 69 µmol/L) with normal preoperative levels in three cases without associated metabolic decompensation. There were two cases of perioperative hyperammonemia (72 and 69 µmol/L) secondary to preoperative metabolic decompensation (137 and 92 µmol/L) with the levels progressively dropping and normalizing in the first 24-48 hours, respectively. CONCLUSIONS: Procedures under anesthesia on pediatric patients with urea cycle diseases should be performed by experienced multidisciplinary teams at specialized centers. Perioperative management focused on avoiding catabolism (especially during fasting) and monitoring signs associated with metabolic decompensation to allow for its early treatment should be included in routine anesthetic techniques for children with urea cycle disorders.


Subject(s)
Hyperammonemia , Urea Cycle Disorders, Inborn , Ammonia , Anesthesia, General , Child , Humans , Prognosis , Urea Cycle Disorders, Inborn/complications
19.
Soft Matter ; 16(11): 2725-2735, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32115597

ABSTRACT

Transmembrane pH gradient poly(isoprene)-block-poly(ethylene glycol) (PI-b-PEG) polymersomes were investigated for their potential use in the detoxification of ammonia, a metabolite that is excessively present in patients suffering from urea cycle disorders and advanced liver diseases, and which causes neurotoxic effects (e.g., hepatic encephalopathy). Polymers varying in PI and PEG block length were synthesized via nitroxide-mediated polymerization and screened for their ability to self-assemble into polymersomes in aqueous media. Ammonia sequestration by the polymersomes was investigated in vitro. While most vesicular systems were able to capture ammonia in simulated intestinal fluids, uptake was lost in partially dehydrated medium mimicking conditions in the colon. Polymeric crosslinking of residual olefinic bonds in the PI block increased polymersome stability, partially preserving the ammonia capture capacity in the simulated colon environment. These more stable vesicular systems hold promise for the chronic oral treatment of hyperammonemia.


Subject(s)
Ammonia/chemistry , Drug Carriers/chemistry , Hepatic Encephalopathy/drug therapy , Inactivation, Metabolic/genetics , Ammonia/metabolism , Butadienes/chemistry , Butadienes/pharmacology , Drug Carriers/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Hemiterpenes/chemistry , Hemiterpenes/pharmacology , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/metabolism , Humans , Hydrogen-Ion Concentration , Liver Diseases/complications , Liver Diseases/drug therapy , Liver Diseases/metabolism , Methacrylates/chemistry , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polymerization , Polymers/chemistry , Polymers/pharmacology , Proton-Motive Force/drug effects , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism , Water/metabolism
20.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990680

ABSTRACT

BACKGROUNDLiver disease in urea cycle disorders (UCDs) ranges from hepatomegaly and chronic hepatocellular injury to cirrhosis and end-stage liver disease. However, the prevalence and underlying mechanisms are unclear.METHODSWe estimated the prevalence of chronic hepatocellular injury in UCDs using data from a multicenter, longitudinal, natural history study. We also used ultrasound with shear wave elastography and FibroTest to evaluate liver stiffness and markers of fibrosis in individuals with argininosuccinate lyase deficiency (ASLD), a disorder with high prevalence of elevated serum alanine aminotransferase (ALT). To understand the human observations, we evaluated the hepatic phenotype of the AslNeo/Neo mouse model of ASLD.RESULTSWe demonstrate a high prevalence of elevated ALT in ASLD (37%). Hyperammonemia and use of nitrogen-scavenging agents, 2 markers of disease severity, were significantly (P < 0.001 and P = 0.001, respectively) associated with elevated ALT in ASLD. In addition, ultrasound with shear wave elastography and FibroTest revealed increased echogenicity and liver stiffness, even in individuals with ASLD and normal aminotransferases. The AslNeo/Neo mice mimic the human disorder with hepatomegaly, elevated aminotransferases, and excessive hepatic glycogen noted before death (3-5 weeks of age). This excessive hepatic glycogen is associated with impaired hepatic glycogenolysis and decreased glycogen phosphorylase and is rescued with helper-dependent adenovirus expressing Asl using a liver-specific (ApoE) promoter.CONCLUSIONOur results link urea cycle dysfunction and impaired hepatic glucose metabolism and identify a mouse model of liver disease in the setting of urea cycle dysfunction.TRIAL REGISTRATIONThis study has been registered at ClinicalTrials.gov (NCT03721367, NCT00237315).FUNDINGFunding was provided by NIH, Burroughs Wellcome Fund, NUCDF, Genzyme/ACMG Foundation, and CPRIT.


Subject(s)
Argininosuccinate Lyase/metabolism , Liver Diseases/metabolism , Liver Glycogen/metabolism , Alanine Transaminase/blood , Animals , Chronic Disease , Disease Models, Animal , Humans , Liver Diseases/complications , Liver Diseases/enzymology , Longitudinal Studies , Mice , Urea Cycle Disorders, Inborn/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...