Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
Add more filters

Publication year range
1.
Br J Haematol ; 187(3): 386-395, 2019 11.
Article in English | MEDLINE | ID: mdl-31273765

ABSTRACT

Hereditary spherocytosis (HS) is characterised by increased osmotic fragility and enhanced membrane loss of red blood cells (RBC) due to defective membrane protein complexes. In our diagnostic laboratory, we observed that pyruvate kinase (PK) activity in HS was merely slightly elevated with respect to the amount of reticulocytosis. In order to evaluate whether impaired PK activity is a feature of HS, we retrospectively analysed laboratory data sets from 172 unrelated patients with HS, hereditary elliptocytosis (HE), glucose-6-phosphate dehydrogenase (G6PD) or PK deficiency, sickle cell or haemoglobin C disease, or ß-thalassaemia minor. Results from linear regression analysis provided proof that PK activity decreases with rising reticulocyte counts in HS (R2  = 0·15; slope = 9·09) and, less significantly, in HE (R2  = 0·021; slope = 8·92) when compared with other haemolytic disorders (R2  ≥ 0·65; slopes ≥ 78·6). Reticulocyte-adjusted erythrocyte PK activity levels were significantly lower in HS and even declined with increasing reticulocytes (R2  = 0·48; slope = -9·74). In this report, we describe a novel association between HS and decreased PK activity that is apparently caused by loss of membrane-bound PK due to impaired structural integrity of the RBC membrane and may aggravate severity of haemolysis in HS.


Subject(s)
Erythrocyte Membrane/enzymology , Erythrocytes, Abnormal/enzymology , Pyruvate Kinase/metabolism , Spherocytosis, Hereditary/enzymology , Adolescent , Adult , Aged , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/pathology , Child , Child, Preschool , Erythrocyte Membrane/pathology , Erythrocytes, Abnormal/pathology , Female , Hemoglobin C Disease/enzymology , Hemoglobin C Disease/pathology , Humans , Infant , Male , Middle Aged , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/pathology , Reticulocytes/enzymology , Reticulocytes/pathology , Spherocytosis, Hereditary/pathology , beta-Thalassemia/enzymology , beta-Thalassemia/pathology
2.
J Inherit Metab Dis ; 42(5): 839-849, 2019 09.
Article in English | MEDLINE | ID: mdl-31111503

ABSTRACT

Triosephosphate isomerase (TPI) deficiency is a fatal genetic disorder characterized by hemolytic anemia and neurological dysfunction. Although the enzyme defect in TPI was discovered in the 1960s, the exact etiology of the disease is still debated. Some aspects indicate the disease could be caused by insufficient enzyme activity, whereas other observations indicate it could be a protein misfolding disease with tissue-specific differences in TPI activity. We generated a mouse model in which exchange of a conserved catalytic amino acid residue (isoleucine to valine, Ile170Val) reduces TPI specific activity without affecting the stability of the protein dimer. TPIIle170Val/Ile170Val mice exhibit an approximately 85% reduction in TPI activity consistently across all examined tissues, which is a stronger average, but more consistent, activity decline than observed in patients or symptomatic mouse models that carry structural defect mutant alleles. While monitoring protein expression levels revealed no evidence for protein instability, metabolite quantification indicated that glycolysis is affected by the active site mutation. TPIIle170Val/Ile170Val mice develop normally and show none of the disease symptoms associated with TPI deficiency. Therefore, without the stability defect that affects TPI activity in a tissue-specific manner, a strong decline in TPI catalytic activity is not sufficient to explain the pathological onset of TPI deficiency.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Carbohydrate Metabolism, Inborn Errors/pathology , Catalytic Domain/genetics , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Animals , Behavior, Animal , Carbohydrate Metabolism, Inborn Errors/enzymology , Disease Models, Animal , Enzyme Stability , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Protein Multimerization
3.
Biochim Biophys Acta Gen Subj ; 1862(6): 1401-1409, 2018 06.
Article in English | MEDLINE | ID: mdl-29571745

ABSTRACT

Human triosephosphate isomerase (TIM) deficiency is a very rare disease, but there are several mutations reported to be causing the illness. In this work, we produced nine recombinant human triosephosphate isomerases which have the mutations reported to produce TIM deficiency. These enzymes were characterized biophysically and biochemically to determine their kinetic and stability parameters, and also to substitute TIM activity in supporting the growth of an Escherichia coli strain lacking the tim gene. Our results allowed us to rate the deleteriousness of the human TIM mutants based on the type and severity of the alterations observed, to classify four "unknown severity mutants" with altered residues in positions 62, 72, 122 and 154 and to explain in structural terms the mutation V231M, the most affected mutant from the kinetic point of view and the only homozygous mutation reported besides E104D.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Carbohydrate Metabolism, Inborn Errors/enzymology , Mutation , Triose-Phosphate Isomerase/chemistry , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Carbohydrate Metabolism, Inborn Errors/genetics , Enzyme Stability , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Triose-Phosphate Isomerase/genetics
4.
Eur J Haematol ; 98(6): 584-589, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28295642

ABSTRACT

OBJECTIVE: Here, we present a 7-year-old patient suffering from severe haemolytic anaemia. The most common cause of chronic hereditary non-spherocytic haemolytic anaemia is red blood cell pyruvate kinase (PK-R) deficiency. Because red blood cells rely solely on glycolysis to generate ATP, PK-R deficiency can severely impact energy supply and cause reduction in red blood cell lifespan. We determined the underlying cause of the anaemia and investigated how erythroid precursors in the patient survive. METHODS: PK activity assays, Western blot and Sanger sequencing were employed to determine the underlying cause of the anaemia. Patient erythroblasts were cultured and reticulocytes were isolated to determine PK-R and PKM2 contribution to glycolytic activity during erythrocyte development. RESULTS: We found a novel homozygous mutation (c.583G>A) in the PK-R coding gene (PKLR). Although this mutation did not influence PKLR mRNA production, no PK-R protein could be detected in the red blood cells nor in its precursors. In spite of the absence of PK-R, the reticulocytes of the patient exhibited 20% PK activity compared with control. Western blotting revealed that patient erythroid precursors, like controls, express residual PKM2. CONCLUSIONS: We conclude that PKM2 rescues glycolysis in PK-R-deficient erythroid precursors.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Carrier Proteins/genetics , Erythroblasts/enzymology , Membrane Proteins/genetics , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/genetics , Reticulocytes/enzymology , Thyroid Hormones/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Base Sequence , Cell Differentiation , Child , Consanguinity , Erythroblasts/pathology , Gene Expression , Glycolysis/genetics , Homozygote , Humans , Male , Membrane Proteins/deficiency , Mutation , Myeloid Cells/cytology , Myeloid Cells/enzymology , Primary Cell Culture , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reticulocytes/pathology , Thyroid Hormones/deficiency , Thyroid Hormone-Binding Proteins
6.
Biochim Biophys Acta ; 1852(1): 61-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25463631

ABSTRACT

Triosephosphate isomerase (TPI) is a glycolytic enzyme which homodimerizes for full catalytic activity. Mutations of the TPI gene elicit a disease known as TPI Deficiency, a glycolytic enzymopathy noted for its unique severity of neurological symptoms. Evidence suggests that TPI Deficiency pathogenesis may be due to conformational changes of the protein, likely affecting dimerization and protein stability. In this report, we genetically and physically characterize a human disease-associated TPI mutation caused by an I170V substitution. Human TPI(I170V) elicits behavioral abnormalities in Drosophila. An examination of hTPI(I170V) enzyme kinetics revealed this substitution reduced catalytic turnover, while assessments of thermal stability demonstrated an increase in enzyme stability. The crystal structure of the homodimeric I170V mutant reveals changes in the geometry of critical residues within the catalytic pocket. Collectively these data reveal new observations of the structural and kinetic determinants of TPI Deficiency pathology, providing new insights into disease pathogenesis.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Carbohydrate Metabolism, Inborn Errors/pathology , Catalytic Domain , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Animals , Behavior, Animal , Carbohydrate Metabolism, Inborn Errors/enzymology , Disease Models, Animal , Drosophila , Enzyme Stability , Humans , Mutation , Triose-Phosphate Isomerase/chemistry , Triose-Phosphate Isomerase/genetics
7.
Blood Cells Mol Dis ; 57: 100-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26832193

ABSTRACT

Pyruvate kinase (PK) deficiency is known as being the most common cause of chronic nonspherocytic hemolytic anemia (CNSHA). Clinical PK deficiency is transmitted as an autosomal recessive trait, that can segregate neither in homozygous or in a compound heterozygous modality, respectively. Two PK genes are present in mammals: the pyruvate kinase liver and red blood cells (PK-LR) and the pyruvate kinase muscle (PK-M), of which only the first encodes for the isoenzymes normally expressed in the red blood cells (R-type) and in the liver (L-type). Several reports have been published describing a large variety of genetic defects in PK-LR gene associated to CNSHA. Herein, we present a review of about 250 published mutations and six polymorphisms in PK-LR gene with the corresponding clinical and molecular data. We consulted the PubMed website for searching mutations and papers, along with two main databases: the Leiden Open Variation Database (LOVD, https://grenada.lumc.nl/LOVD2/mendelian_genes/home.php?select_db=PKLR) and Human Gene Mutation Database (HGMD, http://www.hgmd.cf.ac.uk/ac/gene.php?gene=PKLR) for selecting, reviewing and listing the annotated PK-LR gene mutations present in literature. This paper is aimed to provide useful information to clinicians and laboratory professionals regarding overall reported PK-LR gene mutations, also giving the opportunity to harmonize data regarding PK-deficient individuals.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Databases, Genetic , Erythrocytes/enzymology , Mutation , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Animals , Erythrocytes/pathology , Genes, Recessive , Heterozygote , Homozygote , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/enzymology , Liver/pathology , Muscles/enzymology , Muscles/pathology , Polymorphism, Genetic , Pyruvate Kinase/metabolism , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/pathology
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 33(1): 53-6, 2016 Feb.
Article in Zh | MEDLINE | ID: mdl-26829734

ABSTRACT

OBJECTIVE: To evaluate the feasibility of genetic and prenatal diagnosis for a family affected with pyruvate kinase deficiency (PKD). METHODS: Targeted sequence capture and high-throughput sequencing technology was used to detect the exons and exon-intron boundaries of the PKLR gene in a clinically suspected PKD patient. Meanwhile, the genotype of the pedigree was validated by Sanger sequencing. Prenatal genetic diagnosis was performed by amniotic fluid sampling after genotype of the mother of the proband was determined. RESULTS: The proband was found to harbor double heterozygous mutations, c.661G>A (Asp221Asn) and c.1528C>T (Arg510Ter), which resulted in amino acid substitution Asp221Asn and Arg510Ter. Such mutations were confirmed by Sanger sequencing. The mother and father of the proband were detected to have respectively carried c.1528C>T (Arg510Ter) and c.661G>A (Asp221Asn) mutation. The fetus was found to have carried the same mutations as the proband. Following selected abortion, analysis of fetal tissue was consistent with the result of prenatal diagnosis. CONCLUSION: The compound mutations of c.661G>A and c.1528C>T of PKLR gene probably underlie the PKD in the family. Prenatal diagnosis of the mutations analysis can facilitate detection of affected fetus in time.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/genetics , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/embryology , Base Sequence , Child, Preschool , DNA Mutational Analysis , Exons , Female , Genotype , Humans , Male , Molecular Sequence Data , Mutation , Pedigree , Pregnancy , Prenatal Diagnosis , Pyruvate Kinase/metabolism , Pyruvate Metabolism, Inborn Errors/embryology
9.
J Cell Sci ; 126(Pt 14): 3151-8, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23641070

ABSTRACT

Triosephosphate isomerase (TPI) is a glycolytic enzyme that converts dihydroxyacetone phosphate (DHAP) into glyceraldehyde 3-phosphate (GAP). Glycolytic enzyme dysfunction leads to metabolic diseases collectively known as glycolytic enzymopathies. Of these enzymopathies, TPI deficiency is unique in the severity of neurological symptoms. The Drosophila sugarkill mutant closely models TPI deficiency and encodes a protein prematurely degraded by the proteasome. This led us to question whether enzyme catalytic activity was crucial to the pathogenesis of TPI sugarkill neurological phenotypes. To study TPI deficiency in vivo we developed a genomic engineering system for the TPI locus that enables the efficient generation of novel TPI genetic variants. Using this system we demonstrate that TPI sugarkill can be genetically complemented by TPI encoding a catalytically inactive enzyme. Furthermore, our results demonstrate a non-metabolic function for TPI, the loss of which contributes significantly to the neurological dysfunction in this animal model.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Carbohydrate Metabolism, Inborn Errors/enzymology , Drosophila melanogaster/physiology , Longevity , Paralysis/enzymology , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Animals , Carbohydrate Metabolism, Inborn Errors/genetics , Catalysis , Dihydroxyacetone Phosphate/metabolism , Disease Models, Animal , Drosophila melanogaster/enzymology , Female , Gene Knockout Techniques , Genetic Complementation Test , Genetic Engineering , Glyceraldehyde 3-Phosphate/metabolism , Glycolysis/genetics , Hot Temperature/adverse effects , Male , Mutation/genetics , Paralysis/genetics , Stress, Physiological/genetics , Transgenes/genetics , Triose-Phosphate Isomerase/genetics
10.
Am J Hematol ; 90(9): 825-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26087744

ABSTRACT

Over the last several decades, our understanding of the genetic variation, pathophysiology, and complications of the hemolytic anemia associated with red cell pyruvate kinase deficiency (PKD) has expanded. Nonetheless, there remain significant gaps in our knowledge with regard to clinical care and monitoring. Treatment remains supportive with phototherapy and/or exchange transfusion in the newborn period, regular or intermittent red cell transfusions in children and adults, and splenectomy to decrease transfusion requirements and/or anemia related symptoms. In this article, we review the clinical diversity of PKD, the current standard of treatment and for supportive care, the complications observed, and future treatment directions.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Erythrocyte Transfusion , Exchange Transfusion, Whole Blood , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/therapy , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Anemia, Hemolytic, Congenital Nonspherocytic/surgery , Child , Disease Management , Humans , Infant, Newborn , Phototherapy , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/pathology , Pyruvate Metabolism, Inborn Errors/surgery , Splenectomy
11.
Biochim Biophys Acta ; 1834(12): 2702-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056040

ABSTRACT

The deficiency of human triosephosphate isomerase (HsTIM) generates neurological alterations, cardiomyopathy and premature death. The mutation E104D is the most frequent cause of the disease. Although the wild type and mutant exhibit similar kinetic parameters, it has been shown that the E104D substitution induces perturbation of an interfacial water network that, in turn, reduces the association constant between subunits promoting enzyme inactivation. To gain further insight into the effects of the mutation on the structure, stability and function of the enzyme, we measured the sensitivity of recombinant E104D mutant and wild type HsTIM to limited proteolysis. The mutation increases the susceptibility to proteolysis as consequence of the loss of rigidity of its overall 3-D structure. Unexpectedly, it was observed that proteolysis of wild type HsTIM generated two different stable nicked dimers. One was formed in relatively short times of incubation with proteinase K; as shown by spectrometric and crystallographic data, it corresponded to a dimer containing a nicked monomer and an intact monomer. The formation of the other nicked species requires relatively long incubation times with proteinase K and corresponds to a dimer with two clipped subunits. The first species retains 50% of the original activity, whereas the second species is inactive. Collectively, we found that the E104D mutant is highly susceptible to proteolysis, which in all likelihood contributes to the pathogenesis of enzymopathy. In addition, the proteolysis data on wild type HsTIM illustrate an asymmetric conduct of the two monomers.


Subject(s)
Amino Acid Substitution , Mutation, Missense , Protein Multimerization , Proteolysis , Triose-Phosphate Isomerase/chemistry , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Carbohydrate Metabolism, Inborn Errors/enzymology , Carbohydrate Metabolism, Inborn Errors/genetics , Enzyme Stability/genetics , Humans , Protein Structure, Quaternary , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/genetics , Triose-Phosphate Isomerase/metabolism
12.
Lik Sprava ; (7-8): 73-7, 2011.
Article in Russian | MEDLINE | ID: mdl-22768742

ABSTRACT

Erythrocyte enzymes participate in the main interactions promoting utilization of glucose-glycolytic, pentosophosphate cycles and glutation system. In this report we study on erythrocyte G6PD deficiency which is the impairment related to the gender and expressed with development of acute drug-associated hemolytic anemia. Out of 13187 studied subjects 122 showed carrying of deficiency of erythrocyte G6PD activity, from them 98 (80.3%) subjects were male, and 24 (19.7%) female. As a whole, among the revealed in the population studies, and also verified in clinic of the persons with deficiency of erythrocyte G6PD there were marked different pathological phenotypes: hereditary nonspherecytary hemolytic anemia, acute drug-induced hemolytic anemia, asymptomatic gene carrying and, selected by us disease with few symptoms. As a whole, among the revealed in the population studies, and also verified in clinic of the persons with deficiency of erythrocyte G6PD there were marked different pathological phenotypes: hereditary nonspherecytary hemolytic anemia, acute drug-induced hemolytic anemia, asymptomatic gene carrying and, selected by us disease with few symptoms.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Anemia, Hemolytic/genetics , Ethnicity , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase/genetics , Alleles , Anemia, Hemolytic/enzymology , Anemia, Hemolytic/ethnology , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/ethnology , Carrier State , Erythrocytes/drug effects , Erythrocytes/enzymology , Female , Gene Frequency , Genotype , Glucosephosphate Dehydrogenase Deficiency/enzymology , Glucosephosphate Dehydrogenase Deficiency/ethnology , Hemolysis/drug effects , Humans , Male , Oxidants/adverse effects , Phenotype , Prescription Drugs/adverse effects , Prevalence , Uzbekistan/epidemiology
13.
Biochim Biophys Acta ; 1792(12): 1168-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19786097

ABSTRACT

The triosephosphate isomerase (TPI) functions at a metabolic cross-road ensuring the rapid equilibration of the triosephosphates produced by aldolase in glycolysis, which is interconnected to lipid metabolism, to glycerol-3-phosphate shuttle and to the pentose phosphate pathway. The enzyme is a stable homodimer, which is catalytically active only in its dimeric form. TPI deficiency is an autosomal recessive multisystem genetic disease coupled with hemolytic anemia and neurological disorder frequently leading to death in early childhood. Various genetic mutations of this enzyme have been identified; the mutations result in decrease in the catalytic activity and/or the dissociation of the dimers into inactive monomers. The impairment of TPI activity apparently does not affect the energy metabolism at system level; however, it results in accumulation of dihydroxyacetone phosphate followed by its chemical conversion into the toxic methylglyoxal, leading to the formation of advanced glycation end products. By now, the research on this disease seems to enter a progressive stage by adapting new model systems such as Drosophila, yeast strains and TPI-deficient mouse, which have complemented the results obtained by prediction and experiments with recombinant proteins or erythrocytes, and added novel data concerning the complexity of the intracellular behavior of mutant TPIs. This paper reviews the recent studies on the structural and catalytic changes caused by mutation and/or nitrotyrosination of the isomerase leading to the formation of an aggregation-prone protein, a characteristic of conformational disorders.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Triose-Phosphate Isomerase/deficiency , Animals , Animals, Genetically Modified , Drosophila/genetics , Mice/genetics
14.
Science ; 195(4280): 783-5, 1977 Feb 25.
Article in English | MEDLINE | ID: mdl-836588

ABSTRACT

Hereditary hemolytic anemia, a dominantly transmitted disorder, has affected 12 family members spanning three generations. The concentration of adenosine triphosphate in the red cells was about half that of comparably reticulocyte-rich blood. Since adenosine deaminase and adenosine kinase compete for a common substrate, the greatly increased activity of the former may interfere with nucleotide salvage via the latter.


Subject(s)
Adenosine Deaminase/blood , Adenosine Triphosphate/blood , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Erythrocytes/enzymology , Nucleoside Deaminases/blood , Adenine Nucleotides/blood , Adenosine Kinase/blood , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Genes, Dominant , Humans , Pedigree , Reticulocytes/metabolism
15.
Indian J Pediatr ; 86(8): 692-699, 2019 08.
Article in English | MEDLINE | ID: mdl-31030358

ABSTRACT

OBJECTIVES: Glucose-6-phosphate isomerase (GPI) deficiency is an autosomal recessive genetic disorder causing hereditary non-spherocytic hemolytic anemia (HNSHA) coupled with a neurological disorder. The aim of this study was to identify GPI genetic defects in a cohort of Indian patients with HNSHA coupled with neurological dysfunction. METHODS: Thirty-five patients were screened for GPI deficiency in the HNSHA patient group; some were having neurological dysfunction. Enzyme activity was measured by spectrophotometric method. The genetic study was done by single-stranded conformation polymorphism (SSCP) analysis, restriction fragment length polymorphism (RFLP) analysis by the restriction enzyme AciI for p.Arg347His (p.R347H) and confirmation by Sanger's sequencing. RESULTS: Out of 35 patients, 15 showed 35% to 70% loss of GPI activity, leading to neurological problems with HNSHA. Genetic analysis of PCR products of exon 12 of the GPI gene showed altered mobility on SSCP gel. Sanger's sequencing revealed a homozygous c1040G > A mutation predicting a p.Arg347His replacement which abolishes AciI restriction site. The molecular modeling analysis suggests p.Arg347 is involved in dimerization of the enzyme. Also, this mutation generates a more labile enzyme which alters its three-dimensional structure and function. CONCLUSIONS: This report describes the high prevalence of p.Arg347His pathogenic variant identified in Indian GPI deficient patients with hemolytic anemia and neuromuscular impairment. It suggests that neuromuscular impairment with hemolytic anemia cases could be investigated for p.Arg347His pathogenic variant causing GPI deficiency because of neuroleukin activity present in the GPI monomer which has neuroleukin action at the same active site and generates neuromuscular problems as well as hemolytic anemia.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Glucose-6-Phosphate Isomerase/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Neuromuscular Diseases/enzymology , Neuromuscular Diseases/genetics , Adolescent , Child , Child, Preschool , Female , Humans , India , Infant , Male , Mutation, Missense , Prevalence
16.
J Clin Pathol ; 72(6): 393-398, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30918013

ABSTRACT

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


Subject(s)
Adenylate Kinase/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Erythrocytes/enzymology , Mutation, Missense , Adenylate Kinase/blood , Adenylate Kinase/chemistry , Adenylate Kinase/deficiency , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Child , DNA Mutational Analysis/methods , Female , Genetic Predisposition to Disease , Heredity , High-Throughput Nucleotide Sequencing , Humans , India , Male , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship
17.
Exp Hematol ; 35(8): 1182-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17662886

ABSTRACT

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


Subject(s)
Adenylate Kinase/deficiency , Adenylate Kinase/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Erythrocytes/enzymology , Mutation , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/blood , Adenylate Kinase/chemistry , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Circular Dichroism , Enzyme Stability , Escherichia coli/genetics , Frameshift Mutation , Humans , Kinetics , Models, Molecular , Mutation, Missense , Polymerase Chain Reaction , Protein Conformation , RNA/genetics , RNA/isolation & purification , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Restriction Mapping , Sequence Deletion
18.
J Clin Invest ; 62(2): 294-301, 1978 Aug.
Article in English | MEDLINE | ID: mdl-27532

ABSTRACT

In the erythrocytes of a patient with hereditary nonspherocytic hemolytic anemia, a homozygous expression of hexokinase deficiency was detected. The mutant enzyme was characterized by normal kinetic parameters with respect to its substrates, glucose and MgATP2-, normal pH optimum, normal heat stability at 40 degrees C, but abnormal behavior with respect to its regulation by glucose-1,6-diphosphate and inorganic phosphate, and an altered electrophoretic pattern. Interpretation of the results revealed the presence of two different hexokinases type I in normal human erythrocytes: one enzyme with a high affinity for glucose-1,6-diphosphate, the inhibition of which is regulated by inorganic phosphate; and another enzyme with a lower affinity for the inhibitor, not regulated by inorganic phosphate. The former enzyme was not detectable in the erythrocytes of the patient, whereas the presence of the latter enzyme could be demonstrated.


Subject(s)
Erythrocytes/enzymology , Hexokinase/deficiency , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Electrophoresis, Disc , Female , Glucosephosphates/pharmacology , Hexokinase/blood , Hexokinase/genetics , Hot Temperature , Humans , Hydrogen-Ion Concentration , Kinetics , Male , Mutation , Phosphates/pharmacology
19.
J Clin Invest ; 95(4): 1702-9, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7706479

ABSTRACT

DNA analysis was performed on 30 unrelated patients with hereditary nonspherocytic hemolytic anemia (HNSHA) who had been found to be pyruvate kinase (PK) deficient by enzyme assay. 19 different mutations were identified among 58 of the 60 alleles at risk. 13 of these were missense mutations that caused single amino acid changes. Included were the following nucleotide substitutions: 401A, 464C, 993A, 1022C, 1076A, 1178G, 1179A, 1373A, 1378A, 1456T, 1484T, 1493A, 1529A. The remaining six mutations were as follows: two nonsense mutations, 721T and 808T; a nucleotide deletion, 307C; a nucleotide insertion, 1089GG; a three nucleotide in frame deletion, 391-392-393 and a deletion of 1149 bp from the PKLR gene that resulted in the loss of exon 11. All the patients were studied for two polymorphic sites, nucleotide (nt) 1705 A/C and a microsatellite in intron 11, to better understand the origin of the mutations. The 1529A mutation, which is the most common mutation in the European population, was found in 25 alleles. With a single exception this mutation was in linkage disequilibrium with both of the polymorphic markers, i.e., found with 1705C and 14 repeats in the microsatellite. This finding is consistent with a single origin of this common mutation. Other mutations occurring more than once were of much lower frequency than the 1529A mutation.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Liver/enzymology , Mutation , Polymorphism, Genetic , Pyruvate Kinase/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Base Sequence , Black People/genetics , Conserved Sequence , Crossing Over, Genetic , DNA, Satellite/genetics , Exons/genetics , Humans , Introns/genetics , Linkage Disequilibrium , Models, Genetic , Molecular Sequence Data , Mutagenesis, Insertional , Point Mutation , Population , Pyruvate Kinase/deficiency , Reading Frames/genetics , Sequence Deletion , Species Specificity , White People/genetics
20.
J Clin Invest ; 48(11): 1997-2007, 1969 Nov.
Article in English | MEDLINE | ID: mdl-4980929

ABSTRACT

Analyses of key glycolytic intermediates in freshly drawn red cells from six related individuals suggest that decreased hexokinase activity underlies the hemolytic process in the two members with overt hemolysis. Low red cell glucose 6-phosphate (G6P) was observed not only in the anemic patients but in the presumptive heterozygotes as well and served as a useful marker for the presence of the trait. Hexokinase activity was labile in distilled water hemolysates but was only slightly low when protected by glucose, mercaptoethanol, and ethylenediaminetetraacetate (EDTA). Normal red cell hexokinase was demonstrated to be dependent on glucose for maintenance of activity after heating to 45 degrees C. The cells of the proposita are unable to utilize glucose efficiently at glucose concentrations lower than 0.2 mmole/liter whereas normal cells maintain linear glucose consumption to at least 0.05 mM glucose. These qualitative abnormalities could result from the presence of a mutant hexokinase with an abnormally reactive sulfhydryl group and altered substrate affinity in the red cells of this kindred.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Hexokinase/blood , Adenosine Triphosphate/blood , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Edetic Acid/pharmacology , Erythrocytes/enzymology , Erythrocytes/metabolism , Female , Glucose/metabolism , Glucose/pharmacology , Glucosephosphate Dehydrogenase/blood , Glycolysis , Hemolysis , Heterozygote , Humans , Mercaptoethanol/pharmacology , Sulfhydryl Compounds/blood
SELECTION OF CITATIONS
SEARCH DETAIL