Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Chemother ; 36(3): 179-189, 2024 May.
Article in English | MEDLINE | ID: mdl-37599449

ABSTRACT

Voriconazole (VRC) displays highly variable pharmacokinetics impacting treatment efficacy and safety. To provide evidence for optimizing VRC therapy regimens, the authors set out to determine the factors impacting VRC steady-state trough concentration (Cmin) in patients with various albumin (Alb) level. A total of 275 blood samples of 120 patients and their clinical characteristics and genotypes of CYP2C19, CYP3A4, CYP3A5, CYP2C9, FMO3, ABCB1, POR, NR1I2 and NR1I3 were included in this study. Results of multivariate linear regression analysis demonstrated that C-reactive protein (CRP) and total bilirubin (T-Bil) were predictors of the VRC Cmin adjusted for dose in patients with hypoalbuminemia (Alb < 35 g/L) (R2 = 0.16, P < 0.001). Additionally, in patients with normal albumin level (Alb ≥ 35 g/L), it resulted in a significant model containing factors of the poor metabolizer (PM) CYP2C19 genotype and CRP level (R2 = 0.26, P < 0.001). Therefore, CRP and T-Bil levels ought to receive greater consideration than genetic factors in patients with hypoalbuminemia.


Subject(s)
Antifungal Agents , Hypoalbuminemia , Humans , Voriconazole/adverse effects , Antifungal Agents/adverse effects , Cytochrome P-450 CYP2C19/genetics , Pharmacogenomic Variants , Hypoalbuminemia/genetics , Hypoalbuminemia/chemically induced , Hypoalbuminemia/drug therapy , C-Reactive Protein , Genotype , China
2.
Ann Biol Clin (Paris) ; 81(2): 204-209, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37184254

ABSTRACT

Congenital analbuminemia (CAA) is a very rare genetic disorder characterized by a significant reduced or even complete absence of human serum albumin. Our data describe the clinical features and laboratory results of a case confirmed by mutation analysis of the albumin gene in a 35-year-old man presenting recurrent acute coronary syndrome. To the best of our knowledge, only two cases of coronary artery disease have been reported worldwide without recurrence. Our findings contribute to shed light on the clinical characteristics and biochemical parameters of this disease and confirm that cardiovascular complications must be taken seriously in this pathology. Mutational screening disclosed two novel compound heterozygous nucleotide variations located in intron 12 and in 3'UTR. The prediction of the functional and structural impact of the reported variations using different bioinformatics tools demonstrates that these genetics variations affect RNA transcription and mRNA folding.


Subject(s)
Coronary Thrombosis , Hypoalbuminemia , Male , Humans , Young Adult , Adult , Serum Albumin , Nucleotides , Coronary Thrombosis/complications , Serum Albumin, Human/genetics , Hypoalbuminemia/complications , Hypoalbuminemia/diagnosis , Hypoalbuminemia/genetics , Mutation
3.
Biomolecules ; 13(3)2023 02 22.
Article in English | MEDLINE | ID: mdl-36979342

ABSTRACT

Albumin is a highly abundant plasma protein with multiple functions, including the balance of fluid between body compartments and fatty acid trafficking. Humans with congenital analbuminemia (CAA) do not express albumin due to homozygosity for albumin gene mutation. Lessons about physiological control could be learned from CAA. Remarkably, these patients exhibit an apparently normal lifespan, without substantial impairments in physical functionality. There was speculation that tolerance to albumin deficiency would be characterized by significant upregulation of other plasma proteins to compensate for analbuminemia. It is unknown but possible that changes in plasma protein expression observed in CAA are required for the well-documented survival and general wellness. A systematic review of published case reports was performed to assess plasma protein pattern remodeling in CAA patients who were free of other illnesses that would confound interpretation. From a literature search in Pubmed, Scopus, and Purdue Libraries (updated October 2022), concentration of individual plasma proteins and protein classes were assessed. Total plasma protein concentration was below the reference range in the vast majority of CAA patients in the analysis, as upregulation of other proteins was not sufficient to prevent the decline of total plasma protein when albumin was absent. Nonetheless, an impressive level of evidence in the literature indicated upregulated plasma levels of multiple globulin classes and various specific proteins which may have metabolic functions in common with albumin. The potential role of this altered plasma protein expression pattern in CAA is discussed, and the findings may have implications for other populations with hypoalbuminemia.


Subject(s)
Hypoalbuminemia , Humans , Hypoalbuminemia/genetics , Hypoalbuminemia/congenital , Blood Proteins/genetics , Blood Proteins/metabolism , Albumins , Mutation , Plasma/metabolism
4.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163085

ABSTRACT

The human albumin gene, the most abundant serum protein, is located in the long arm of chromosome 4, near the centromere, position 4q11-3. It is divided by 14 intervening introns into 15 exons, the last of which is untranslated. To date, 74 nucleotide substitutions (mainly missense) have been reported, determining the circulating variants of albumin or pre-albumin. In a heterozygous state, this condition is known as alloalbuminaemia or bisalbuminaemia (OMIM # 103600). The genetic variants are not associated with disease, neither in the heterozygous nor in the homozygous form. Only the variants resulting in familial dysalbuminaemic hyperthyroxinaemia and hypertriiodothyroninaemia are of clinical relevance because affected individuals are at risk of inappropriate treatment or may have adverse drug effects. In 28 other cases, the pathogenic variants (mainly affecting splicing, nonsense, and deletions), mostly in the homozygous form, cause a premature stop in the synthesis of the protein and lead to the condition known as congenital analbuminaemia. In this review, we will summarize the current knowledge of genetic and molecular aspects, functional consequences and potential therapeutic uses of the variants. We will also discuss the molecular defects resulting in congenital analbuminaemia, as well as the biochemical and clinical features of this rare condition.


Subject(s)
Homozygote , Hypoalbuminemia/genetics , Hypoalbuminemia/pathology , Introns , Mutation , Serum Albumin, Human/genetics , Exons , Humans , Hypoalbuminemia/metabolism , Serum Albumin, Human/metabolism
5.
Clin Immunol ; 229: 108776, 2021 08.
Article in English | MEDLINE | ID: mdl-34118401

ABSTRACT

Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH) is a neurodegenerative disorder caused by mutation in the aprataxin (APTX)-coding gene APTX, which is involved in DNA single-strand break repair (SSBR). The neurological abnormalities associated with EAOH are similar to those observed in patients with ataxia-telangiectasia. However, the immunological abnormalities in patients with EAOH have not been described. In this study, we report that EAOH patients have immunological abnormalities, including lymphopenia; decreased levels of CD4+ T-cells, CD8+ T-cells, and B-cells; hypogammaglobulinemia; low T-cell recombination excision circles and kappa-deleting element recombination circles; and oligoclonality of T-cell receptor ß-chain variable repertoire. These immunological abnormalities vary among the EAOH patients. Additionally, mild radiosensitivity in the lymphocytes obtained from the patients with EAOH was demonstrated. These findings suggested that the immunological abnormalities and mild radiosensitivity evident in patients with EAOH could be probably caused by the DNA repair defects.


Subject(s)
Apraxias/immunology , Cerebellar Ataxia/congenital , Hypoalbuminemia/immunology , Adolescent , Adult , Apraxias/genetics , Apraxias/metabolism , Case-Control Studies , Cerebellar Ataxia/genetics , Cerebellar Ataxia/immunology , Cerebellar Ataxia/metabolism , Child , DNA Breaks, Single-Stranded , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , Female , Genes, T-Cell Receptor , Genetic Variation , Humans , Hypoalbuminemia/genetics , Hypoalbuminemia/metabolism , Male , Middle Aged , Mutation , Nuclear Proteins/genetics , Radiation Tolerance/genetics , Radiation Tolerance/immunology , T-Lymphocytes/immunology , Young Adult
6.
Medicine (Baltimore) ; 100(24): e26307, 2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34128868

ABSTRACT

INTRODUCTION: 22q13.3 deletion syndrome is a well-known syndrome characterized by typical clinical findings including neonatal hypotonia, absent or severely delayed speech, intellectual disability, and other various features, and detection of a heterozygous deletion of chromosome 22q13.3 with the involvement of at least part of SHANK3. It is reported that 10% to 29% of patients with 22q13.3 deletion syndrome present lymphedema. Protein-losing enteropathy (PLE) has never been reported in 22q13.3 deletion syndrome. PATIENT CONCERNS: The patient presented to our institution for refractory hypoalbuminemia and chronic lymphedema in both legs. DIAGNOSIS: The patient manifested intellectual disability, absent speech, tooth grinding, dysmorphic face, and abnormal hands and toenails. Copy-number variation sequencing confirmed the maternal deletion in 22q13.31-q13.33 (chr22:46285592-51244566, hg19). The patient was genetically diagnosed with 22q13.3 deletion syndrome. INTERVENTIONS: Low-fat diets and medium-chain triglycerides supplements were prescribed. The patient was recommended to wear compression garments and elevate legs. OUTCOMES: The symptom of diarrhea was resolved, but hypoalbuminemia persisted. Lower extremities lymphedema was gradually becoming severe. CONCLUSIONS: Primary lymphedema and PLE can occur simultaneously in a patient with 22q13.3 deletion syndrome. The 2 phenotypes could share the same genetic etiology of congenital lymphatic abnormalities. CELSR1 deletion may play a role in lymphatic dysplasia. The case also provides additional proof of the pathogenic effect of CELSR1 on hereditary lymphedema.


Subject(s)
Cadherins/genetics , Chromosome Disorders/genetics , Lymphedema/genetics , Protein-Losing Enteropathies/genetics , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , DNA Copy Number Variations , Female , Humans , Hypoalbuminemia/genetics , Intellectual Disability/genetics , Leg/pathology , Young Adult
7.
Sci Rep ; 11(1): 11290, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050200

ABSTRACT

Hypoalbuminemia is associated with vascular endothelial dysfunction and the development of chronic cardiovascular diseases. However, the relationship between serum albumin concentration and blood pressure changes remains controversial. Community-based longitudinal cohort data collected from Korean Genome and Epidemiology Study were used in this study. Hypoalbuminemia was defined as a serum albumin concentration of ≤ 4.0 g/dL. A total of 4325 participants were categorized into control (n = 3157) and hypoalbuminemia (n = 1168) groups. Serum albumin had a non-linear relationship with the risk of hypertension development. A genome-wide association study revealed 71 susceptibility loci associated with hypoalbuminemia. Among susceptibility loci, genetic variations at rs2894536 in LOC107986598 and rs10972486 in ATP8B5P were related to elevated blood pressure. Serum albumin (HR = 0.654, 95% CI 0.521-0.820) and polymorphisms of rs2894536 (HR = 1.176, 95% CI 1.015-1.361) and rs10972486 (HR = 1.152, 95% CI 1.009-1.316) were significant predictors of hypertension development. Increased albumin concentration instrumented by 2 hypoalbuminemia-associated SNPs (rs2894536 and rs10972486) was associated with decreased HRs for hypertension development (HR = 0.762, 95% CI 0.659-0.882 and HR = 0.759, 95% CI 0.656-0.878). Our study demonstrated that genetically determined hypoalbuminemia is a significant predictor of incipient hypertension.


Subject(s)
Hypertension/genetics , Hypoalbuminemia/genetics , Serum Albumin/analysis , Adult , Cohort Studies , Female , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Hypertension/blood , Hypertension/physiopathology , Hypoalbuminemia/blood , Hypoalbuminemia/physiopathology , Longitudinal Studies , Male , Middle Aged , Prognosis , Republic of Korea/epidemiology , Retrospective Studies , Risk Factors
9.
BMC Med Genet ; 21(1): 239, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33261563

ABSTRACT

BACKGROUND: Early-onset chronic diarrhoea often indicates a congenital disorder. Mutation in diacylglycerol o-acyltransferase 1 (DGAT1) has recently been linked to early-onset chronic diarrhoea. To date, only a few cases of DGAT1 deficiency have been reported. Diarrhoea in those cases was severe and developed in the neonatal period or within 2 months after birth. CASE PRESENTATION: Here, we report a female patient with DGAT1 mutations with delayed-onset chronic diarrhoea. The patient had vomiting, hypoalbuminemia, hypertriglyceridemia, and failure to thrive at early infancy. Her intractable chronic diarrhoea occurred until she was 8 months of age. A compound heterozygous DGAT1 mutation was found in the patient, which was first found in the Chinese population. Her symptoms and nutrition status improved after nutritional therapy, including a fat restriction diet. CONCLUSIONS: This case expanded our knowledge of the clinical features of patients with DGAT1 mutations. Intractable diarrhoea with delayed onset could also be a congenital disorder.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Diarrhea/genetics , Failure to Thrive/genetics , Hypertriglyceridemia/genetics , Hypoalbuminemia/genetics , Mutation , Vomiting/genetics , Age of Onset , Base Sequence , Diacylglycerol O-Acyltransferase/deficiency , Diarrhea/diet therapy , Diarrhea/metabolism , Diarrhea/physiopathology , Diet, Fat-Restricted , Failure to Thrive/diet therapy , Failure to Thrive/metabolism , Failure to Thrive/physiopathology , Female , Gene Expression , Heterozygote , Humans , Hypertriglyceridemia/diet therapy , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/physiopathology , Hypoalbuminemia/diet therapy , Hypoalbuminemia/metabolism , Hypoalbuminemia/physiopathology , Infant , Severity of Illness Index , Vomiting/diet therapy , Vomiting/metabolism , Vomiting/physiopathology
10.
Article in English | MEDLINE | ID: mdl-33101765

ABSTRACT

Background: Ataxia with oculomotor apraxia (AOA1) is characterized by early-onset progressive cerebellar ataxia with peripheral neuropathy, oculomotor apraxia and hypoalbuminemia and hypercholesterolemia. Case Report: A 23-year-old previously healthy woman presented with slowly-progressive gait impairment since the age of six years. Neurological examination revealed profound areflexia, chorea, generalized dystonia and oculomotor apraxia. Brain MRI revealed mild cerebellar atrophy and needle EMG showed axonal sensorimotor neuropathy. Whole exome sequencing revealed a mutation in the aprataxin gene. Discussion: AOA1 can present with choreoathetosis mixed with dystonic features, resembling ataxia-telangiectasia. This case is instructive since mixed and complex movement disorders is not very common in AOA1. Highlights: Ataxia with oculomotor apraxia type 1 (AOA1) is characterized by early-onset ataxia and oculomotor apraxia caused by variants in the APTX gene.Ataxia is usually not the sole movement abnormality in AOA1.Hyperkinetic movement disorders, especially chorea and dystonia, may occur.Mixed and complex movement disorders is not very common in AOA1.Patients with early-onset ataxia associated with mixed movement disorders should also be investigated for AOA1.


Subject(s)
Apraxias/physiopathology , Cerebellar Ataxia/congenital , Cerebellum/diagnostic imaging , Chorea/physiopathology , Dystonia/physiopathology , Hypoalbuminemia/physiopathology , Reflex, Abnormal/physiology , Apraxias/diagnostic imaging , Apraxias/genetics , Atrophy , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Cerebellar Diseases/physiopathology , Cerebellum/pathology , DNA-Binding Proteins/genetics , Electromyography , Female , Humans , Hypoalbuminemia/diagnostic imaging , Hypoalbuminemia/genetics , Nuclear Proteins/genetics , Young Adult
11.
Med Clin (Barc) ; 154(7): 275-278, 2020 04 10.
Article in English, Spanish | MEDLINE | ID: mdl-31928748

ABSTRACT

INTRODUCTION AND OBJECTIVES: Congenital analbuminaemia (CCA) (MIM #616000) is an autosomal recessive disorder (prevalence < 1/106) caused by defects in the ALB gene leading to absence or severe reduction of albuminaemia. This paper describes a case of CCA detected and diagnosed in our hospital. MATERIALS AND METHODS: A 42-year old woman showing hypoproteinaemia and hypoalbuminaemia of unknown aetiology. Biochemical study was performed according to routine quality controlled analytical procedures: Albuminaemia (colorimetric and nephelometric methods). Protein electrophoresis (capillary and agarose gel). Molecular study of the ALB gene: DNA extraction, PCR amplification of the 14 coding exons plus adjacent intron regions and Sanger sequencing. RESULTS: After discarding the most common causes of hypoalbuminaemia, the analbuminaemia was confirmed by nephelometry and protein electrophoresis. The proband was found to be homozygous for molecular defect in the ALB gene: variant c.1289+1G>A previously reported as Guimarães variant. CONCLUSIONS: This is the first case of CCA confirmed by molecular study in Spain. The proband shows the Guimarães variant previously described in 4 patients worldwide.


Subject(s)
Hypoalbuminemia , Adult , Exons , Female , Homozygote , Humans , Hypoalbuminemia/genetics , Serum Albumin , Spain
12.
Yonsei Med J ; 60(7): 700-703, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31250585

ABSTRACT

Congenital analbuminemia (CAA) is an autosomal recessive disease characterized by extremely low serum levels of albumin. CAA is caused by various homozygous or heterozygous mutations of the ALB gene. Patients often exhibit no clinical symptoms, aside from rare accompanying conditions, such as fatigue, ankle edema, and hypotension. This case report describes the case of a 28-year-old asymptomatic Korean male referred to our center with hypocalcemia, vitamin D deficiency, and hypoalbuminemia who was diagnosed with CAA. To determine the cause of hypoalbuminemia in the patient, laboratory tests, radiological examination, and DNA sequencing were performed. The patient was confirmed to not exhibit any other clinical conditions that can induce hypoalbuminemia and was diagnosed with CAA using DNA sequencing. The present case of CAA is the first to be reported in Korea.


Subject(s)
Hypoalbuminemia/congenital , Hypoalbuminemia/genetics , Polymorphism, Single Nucleotide , Serum Albumin, Human/genetics , Adult , Humans , Hypoalbuminemia/diagnosis , Hypoalbuminemia/metabolism , Male , Republic of Korea , Sequence Analysis, DNA
13.
Eur J Med Genet ; 62(2): 144-148, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29981851

ABSTRACT

Congenital analbuminemia (OMIM # 616000) is an extremely rare autosomal recessive disorder, caused by variations in the albumin gene (ALB), which is generally thought to be a relatively benign condition in adulthood, but seems to be potentially life threatening in the pre- and peri-natal period. The subject of our study was a consanguineous family, in which we identified two analbuminemic individuals. Mutation analysis of ALB revealed that both are homozygous for a previously unreported insertion in exon 9 (c.1098dupT), causing a subsequent frame-shift with the generation of a premature stop codon, and an aberrant truncated putative protein product, p.Val367fsTer12. This variation is present in heterozygous condition in several other members of the family. The phenotype and the molecular genetics of CAA are discussed.


Subject(s)
Hypoalbuminemia/genetics , Mutation , Serum Albumin, Human/genetics , Adult , Aged , Codon, Terminator , Consanguinity , Female , Humans , Hypoalbuminemia/pathology , Male , Pedigree , Phenotype
14.
Gastroenterology ; 155(1): 130-143.e15, 2018 07.
Article in English | MEDLINE | ID: mdl-29604290

ABSTRACT

BACKGROUND & AIMS: Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS: We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS: In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS: We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Duodenum/metabolism , Fibroblasts/metabolism , Hypoalbuminemia/genetics , Lipid Metabolism Disorders/genetics , Organoids/metabolism , Protein-Losing Enteropathies/genetics , Caco-2 Cells , Case-Control Studies , Caspase 3/metabolism , Caspase 7/metabolism , Child , Child, Preschool , Consanguinity , Dermis/cytology , Diacylglycerol O-Acyltransferase/deficiency , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Netherlands , Phorbols , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Turkey
15.
Article in English | MEDLINE | ID: mdl-28652255

ABSTRACT

Hereditary ataxias are a clinically and genetically heterogeneous family of disorders defined by the inability to control gait and muscle coordination. Given the nonspecific symptoms of many hereditary ataxias, precise diagnosis relies on molecular genetic testing. To this end, we conducted whole-exome sequencing (WES) on a large consanguineous Iranian family with hereditary ataxia and oculomotor apraxia. WES in five affected and six unaffected individuals resulted in the identification of a homozygous novel stop-gain mutation in the APTX gene (c.739A>T; p.Lys247*) that segregates with the phenotype. Mutations in the APTX (OMIM 606350) gene are associated with ataxia with oculomotor apraxia type 1 (OMIM 208920).


Subject(s)
Apraxias/genetics , Cerebellar Ataxia/congenital , DNA-Binding Proteins/genetics , Hypoalbuminemia/genetics , Nuclear Proteins/genetics , Adult , Ataxia/complications , Ataxia/genetics , Cerebellar Ataxia/complications , Cerebellar Ataxia/genetics , Child , Child, Preschool , Consanguinity , DNA-Binding Proteins/metabolism , Female , Humans , Iran , Male , Middle Aged , Mutation/genetics , Nuclear Proteins/metabolism , Pedigree , Phenotype , Spinocerebellar Degenerations/complications , Spinocerebellar Degenerations/genetics
16.
PLoS One ; 12(5): e0177799, 2017.
Article in English | MEDLINE | ID: mdl-28542419

ABSTRACT

Hypoalbuminemia of Hemodialysis (HD) patients is an independent cardiovascular risk factor, however, there is no mechanistic explanation between hypoalbuminemia and vascular injury. In the event of oxidative stress and inflammation to which HD patients are exposed, albumin is oxidized and undetected by common laboratory methods, rendering an apparent hypoalbuminemia. We wanted to show that these circulating modified oxidized albumin molecules cause direct vascular damage, mediating inflammation. Once these in-vivo albumin modifications were reduced in- vitro, the apparent hypoalbuminemia concomitantly with its inflammatory effects, were eliminated. Albumin modification profiles from 14 healthy controls (HC) and 14 HD patients were obtained by mass spectrometry (MS) analyses before and after reduction in- vitro, using redox agent 1,4 dithiothreitol (DTT). Their inflammatory effects were explored by exposing human umbilical endothelial cells (HUVEC) to all these forms of albumin. Albumin separated from hypoalbuminemic HD patients increased endothelial mRNA expression of cytokines and adhesion molecules, and augmented secretion of IL-6. This endothelial inflammatory state was almost fully reverted by exposing HUVEC to the in-vitro reduced HD albumin. MS profile of albumin modifications peaks was similar between HD and HC, but the intensities of the various peaks were significantly different. Abolishing the reversible oxidative modifications by DTT prevented endothelial injury and increased albumin levels. The irreversible modifications such as glycation and sulfonation show low intensities in HD albumin profiles and are nearly unobserved in HC. We showed, for the first time, a mechanistic link between hypoalbuminemia and the pro-inflammatory properties of in-vivo oxidized albumin, initiating vascular injury.


Subject(s)
Hypoalbuminemia/blood , Inflammation Mediators/blood , Serum Albumin/metabolism , Adult , Aged , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Case-Control Studies , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression , Human Umbilical Vein Endothelial Cells , Humans , Hypoalbuminemia/etiology , Hypoalbuminemia/genetics , Male , Middle Aged , Oxidation-Reduction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renal Dialysis/adverse effects , Risk Factors , Serum Albumin/chemistry , Spectrometry, Mass, Electrospray Ionization
17.
Ann Clin Biochem ; 53(Pt 5): 615-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26543026

ABSTRACT

Congenital analbuminaemia is a rare autosomal recessive disorder manifested by the presence of a very low amount of circulating serum albumin. The clinical diagnosis may be challenging because of the absence of unambiguous symptoms and because hypoalbuminemia may have many causes different from a genetic lack of the protein. We describe the clinical and molecular characterization of a new case of congenital analbuminaemia in an infant of apparently non-consanguineous parents from Treves, Germany. For molecular diagnosis, we used our strategy, based on the screening of the albumin gene by single-strand conformation polymorphism, heteroduplex analysis and direct DNA sequencing, which revealed that the proband is homozygous and both parents are heterozygous, for a novel G > T transversion at nucleotide c.270+ 1, the first base of intron 3. The mutation inactivates the strongly conserved GT dinucleotide at the 5' splice site consensus sequence of this intron. In conclusion, we report the clinical findings and the molecular defect of this case, which contributes to a better understanding of the biological mechanism of congenital analbuminaemia.


Subject(s)
Hypoalbuminemia/genetics , Serum Albumin/genetics , Adult , Alternative Splicing , Child, Preschool , DNA Mutational Analysis , Female , Genetic Association Studies , Germany , Humans , Male , Point Mutation , RNA Splice Sites
18.
BMC Med Genet ; 16: 28, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25925991

ABSTRACT

BACKGROUND: Mutations in CCBE1 have been found to be responsible for a subset of families with autosomal recessive Hennekam syndrome. Hennekam syndrome is defined as the combination of generalized lymphatic dysplasia (ie. lymphedema and lymphangiectasia), variable intellectual disability and characteristic dysmorphic features. The patient we describe here has a lymphatic dysplasia without intellectual disability or dysmorphism caused by mutation in CCBE1, highlighting the phenotypic variability that can be seen with abnormalities in this gene. CASE PRESENTATION: Our patient is a 5 week old child of Pakistani descent who presented to our center with generalized edema, ascites, and hypoalbuminemia. She was diagnosed with a protein losing enteropathy secondary to segmental primary intestinal lymphangiectasia. As the generalized edema resolved, it became clear that she had mild persistent lymphedema in her hands and feet. No other abnormalities were noted on examination and development was unremarkable at 27 months of age. Given the suspected genetic etiology and the consanguinity in the family, we used a combination of SNP genotyping and exome sequencing to identify the underlying cause of her disease. We identified several large stretches of homozygosity in the patient that allowed us to sort the variants found in the patient's exome to identify p.C98W in CCBE1 as the likely pathogenic variant. CONCLUSIONS: CCBE1 mutation analysis should be considered in all patients with unexplained lymphatic dysplasia even without the other features of classic Hennekam syndrome.


Subject(s)
Calcium-Binding Proteins/genetics , Craniofacial Abnormalities/genetics , Genital Diseases, Male/genetics , Lymphangiectasis, Intestinal/genetics , Lymphatic System/embryology , Lymphedema/genetics , Tumor Suppressor Proteins/genetics , Consanguinity , DNA Mutational Analysis , Female , Genotype , Humans , Hypoalbuminemia/genetics , Infant , Pakistan , Polydactyly/genetics , Polymorphism, Single Nucleotide , Protein-Losing Enteropathies/genetics
19.
Biochem Med (Zagreb) ; 24(1): 151-8, 2014.
Article in English | MEDLINE | ID: mdl-24627724

ABSTRACT

INTRODUCTION: Congenital analbuminemia is a rare autosomal recessive disorder manifested by the presence of a very low amount of circulating serum albumin. It is an allelic heterogeneous defect, caused by variety of mutations within the albumin gene in homozygous or compound heterozygous state. Herein we report the clinical and molecular characterization of a new case of congenital analbuminemia diagnosed in a female newborn of consanguineous (first degree cousins) parents from Ankara, Turkey, who presented with a low albumin concentration (< 8 g/L) and severe clinical symptoms. MATERIALS AND METHODS: The albumin gene of the index case was screened by single-strand conformation polymorphism, heteroduplex analysis, and direct DNA sequencing. The effect of the splicing mutation was evaluated by examining the cDNA obtained by reverse transcriptase - polymerase chain reaction (RT-PCR) from the albumin mRNA extracted from proband's leukocytes. RESULTS: DNA sequencing revealed that the proband is homozygous, and both parents are heterozygous, for a novel G>A transition at position c.1652+1, the first base of intron 12, which inactivates the strongly conserved GT dinucleotide at the 5' splice site consensus sequence of this intron. The splicing defect results in the complete skipping of the preceding exon (exon 12) and in a frame-shift within exon 13 with a premature stop codon after the translation of three mutant amino acid residues. CONCLUSIONS: Our results confirm the clinical diagnosis of congenital analbuminemia in the proband and the inheritance of the trait and contribute to shed light on the molecular genetics of analbuminemia.


Subject(s)
Alternative Splicing/genetics , Hypoalbuminemia/congenital , Hypoalbuminemia/genetics , Mutation/genetics , Serum Albumin/genetics , Adolescent , Adult , Base Sequence , Female , Heteroduplex Analysis , Humans , Infant, Newborn , Male , Middle Aged , Molecular Sequence Data , Pedigree , Polymorphism, Single-Stranded Conformational , Serum Albumin/deficiency , Serum Albumin, Human , Young Adult
20.
Nature ; 506(7486): 111-5, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24362567

ABSTRACT

Faithful maintenance and propagation of eukaryotic genomes is ensured by three-step DNA ligation reactions used by ATP-dependent DNA ligases. Paradoxically, when DNA ligases encounter nicked DNA structures with abnormal DNA termini, DNA ligase catalytic activity can generate and/or exacerbate DNA damage through abortive ligation that produces chemically adducted, toxic 5'-adenylated (5'-AMP) DNA lesions. Aprataxin (APTX) reverses DNA adenylation but the context for deadenylation repair is unclear. Here we examine the importance of APTX to RNase-H2-dependent excision repair (RER) of a lesion that is very frequently introduced into DNA, a ribonucleotide. We show that ligases generate adenylated 5' ends containing a ribose characteristic of RNase H2 incision. APTX efficiently repairs adenylated RNA-DNA, and acting in an RNA-DNA damage response (RDDR), promotes cellular survival and prevents S-phase checkpoint activation in budding yeast undergoing RER. Structure-function studies of human APTX-RNA-DNA-AMP-Zn complexes define a mechanism for detecting and reversing adenylation at RNA-DNA junctions. This involves A-form RNA binding, proper protein folding and conformational changes, all of which are affected by heritable APTX mutations in ataxia with oculomotor apraxia 1. Together, these results indicate that accumulation of adenylated RNA-DNA may contribute to neurological disease.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Genome, Human/genetics , Nuclear Proteins/metabolism , RNA/metabolism , Adenosine Monophosphate/metabolism , Apraxias/genetics , Ataxia Telangiectasia/genetics , Cell Survival , Cerebellar Ataxia/congenital , DNA/chemistry , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Humans , Hypoalbuminemia/genetics , Models, Molecular , Mutation/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleic Acid Conformation , Protein Conformation , Protein Folding , RNA/chemistry , Ribonuclease H/metabolism , S Phase Cell Cycle Checkpoints , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Structure-Activity Relationship , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...