Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Blood ; 143(10): 866-871, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38118071

ABSTRACT

ABSTRACT: Pyruvate kinase (PK) is a key enzyme in glycolysis, the sole source of adenosine triphosphate, which is essential for all energy-dependent activities of red blood cells. Activating PK shows great potential for treating a broad range of hemolytic anemias beyond PK deficiency, because they also enhance activity of wild-type PK. Motivated by observations of sickle-cell complications in sickle-trait individuals with concomitant PK deficiency, activating endogenous PK offers a novel and promising approach for treating patients with sickle-cell disease.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Sickle Cell , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors , Humans , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Erythrocytes , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/complications
2.
Hematology ; 29(1): 2290746, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38095306

ABSTRACT

Real-world studies of pyruvate kinase (PK) deficiency and estimates of mortality are lacking. This retrospective observational study aimed to identify patients with PK deficiency and compare their overall survival (OS) to that of a matched cohort without PK deficiency. Patients with ≥1 diagnosis code related to PK deficiency were selected from the US Veterans Health Administration (VHA) database (01/1995-07/2019); patients with a physician-documented diagnosis were included (PK deficiency cohort; index: date of first diagnosis code related to PK deficiency). Patients in the PK deficiency cohort were matched 1:5 to patients from the general VHA population (non-PK deficiency cohort; index: random visit date during match's index year). OS from index was compared between the two cohorts. Eighteen patients in the PK deficiency cohort were matched to 90 individuals in the non-PK deficiency cohort (both cohorts: mean age 57 years, 94% males; median follow-up 6.0 and 8.0 years, respectively). At follow-up, patients in the non-PK deficiency cohort had significantly longer OS than the PK deficiency cohort (median OS: 17.1 vs. 10.9 years; hazard ratio: 2.3; p = 0.0306). During their first-year post-index, 75% and 40% of the PK deficiency cohort had laboratory-confirmed anemia and iron overload, respectively. Among patients who died, cause of death was highly heterogeneous. These results highlight the increased risk of mortality and substantial clinical burden among patients with PK deficiency. While the intrinsic characteristics of the VHA database may limit the generalizability of the results, this is the first real-world study to characterize mortality in patients with PK deficiency.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Pyruvate Metabolism, Inborn Errors , Veterans , Male , Humans , Middle Aged , Female , Pyruvate Kinase , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Pyruvate Metabolism, Inborn Errors/diagnosis , Pyruvate Metabolism, Inborn Errors/complications
3.
Blood Rev ; 61: 101103, 2023 09.
Article in English | MEDLINE | ID: mdl-37353463

ABSTRACT

Novel developments in therapies for various hereditary hemolytic anemias reflect the pivotal role of pyruvate kinase (PK), a key enzyme of glycolysis, in red blood cell (RBC) health. Without PK catalyzing one of the final steps of the Embden-Meyerhof pathway, there is no net yield of adenosine triphosphate (ATP) during glycolysis, the sole source of energy production required for proper RBC function and survival. In hereditary hemolytic anemias, RBC health is compromised and therefore lifespan is shortened. Although our knowledge on glycolysis in general and PK function in particular is solid, recent advances in genetic, molecular, biochemical, and metabolic aspects of hereditary anemias have improved our understanding of these diseases. These advances provide a rationale for targeting PK as therapeutic option in hereditary hemolytic anemias other than PK deficiency. This review summarizes the knowledge, rationale, (pre)clinical trials, and future advances of PK activators for this important group of rare diseases.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Hemolytic, Congenital , Anemia, Hemolytic , Humans , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Anemia, Hemolytic/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Erythrocytes/metabolism , Anemia, Hemolytic, Congenital/therapy , Anemia, Hemolytic, Congenital/metabolism
4.
Eur J Haematol ; 106(4): 484-492, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33370479

ABSTRACT

OBJECTIVES: Pyruvate kinase (PK) deficiency is caused by PKLR gene mutations, leading to defective red blood cell glycolysis and hemolytic anemia. Rates of comorbidities and complications by transfusion history and relative to the general population remain poorly quantified. METHODS: Data for patients aged ≥ 18 years with two confirmed PKLR mutations were obtained from the PK deficiency Natural History Study (NCT02053480). Frequencies of select conditions were compared with an age- and sex-matched cohort from a general insured US population without PK deficiency. RESULTS: Compared with the matched population (n = 1220), patients with PK deficiency (n = 122) had significantly higher lifetime rates of osteoporosis, liver cirrhosis, and pulmonary hypertension; splenectomy and cholecystectomy rates were also significantly higher in the 8 years before the index date. Sixty-five (53.3%) patients with PK deficiency were classified as regularly transfused, 30 (24.6%) as occasionally transfused, and 27 (22.1%) as never transfused. Regularly transfused patients were significantly more likely than never transfused patients to have had splenectomy, cholecystectomy, and/or thrombosis. Liver iron overload was reported in 62% of patients and occurred regardless of transfusion cohort. CONCLUSIONS: Even never transfused patients with PK deficiency had higher rates of select comorbidities and complications than individuals without PK deficiency.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/epidemiology , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/epidemiology , Adult , Alleles , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Comorbidity , Female , Genotype , Humans , Male , Middle Aged , Mutation , Prevalence , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/etiology , Young Adult
5.
Eur J Haematol ; 105(2): 173-184, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32279356

ABSTRACT

OBJECTIVES: Pyruvate kinase deficiency (PK deficiency) is a rare disorder caused by compound heterozygosity or homozygosity for > 300 mutations in the PKLR gene. To understand PK deficiency prevalence, we conducted a systematic literature review. METHODS: We queried Embase and Medline for peer-reviewed references reporting PK deficiency prevalence/incidence, PKLR mutant allele frequency (MAF) among the general population, or crude results from which these metrics could be derived. RESULTS: Of 1390 references screened, 1296 were excluded after title/abstract review; 60 were excluded after full-text review. Four of the remaining 34 studies were considered high-quality for estimating PK deficiency prevalence. Two high-quality studies identified cases from source populations of known sizes, producing estimates of diagnosed PK deficiency prevalence of 3.2 and 8.5 per million. Another high-quality study derived an estimate of diagnosed PK deficiency prevalence of 6.5 per million by screening jaundiced newborns. The final high-quality study estimated total diagnosed and undiagnosed PK deficiency prevalence to be 51 per million through extrapolation from observed MAFs. CONCLUSIONS: We conclude that prevalence of clinically diagnosed PK deficiency is likely between 3.2 and 8.5 per million in Western populations, while the prevalence of diagnosed and undiagnosed PK deficiency could possibly be as high as 51 per million.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/epidemiology , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/epidemiology , Alleles , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Mutation , Population Surveillance , Prevalence , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/etiology
6.
Hemoglobin ; 43(4-5): 292-295, 2019.
Article in English | MEDLINE | ID: mdl-31645145

ABSTRACT

In this study, we report on a compound heterozygote for variants in the key erythroid transcription factor Krüppel-like factor 1 (KLF1) gene in a patient who presented with severe, transfusion-dependent hemolytic anemia. The red cells were normochromic and normocytic, and resembled those seen in patients with congenital nonspherocytic hemolytic anemia (CNSHA). Next generation sequencing (NGS) revealed that the patient was a compound heterozygote for the KLF1 frameshift variant c.519_525dup (p.Gly176ArgfsTer179) and a missense variant c.1012C>A (p.Pro338Thr). This report adds to the wide clinical spectrum of KLF1 gene variants. We suggest that loss of KLF1 should be considered in otherwise unexplained cases of congenital hemolytic anemia.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Genetic Variation , Kruppel-Like Transcription Factors/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Frameshift Mutation , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Mutation, Missense , Pedigree
7.
Pediatr Clin North Am ; 65(3): 579-595, 2018 06.
Article in English | MEDLINE | ID: mdl-29803284

ABSTRACT

Mature red blood cells are reliant on the glycolytic pathway for energy production and the hexose monophosphate shunt for cell protection from oxidative insults. The most common red blood cell enzyme disorders are characterized by hemolysis but with wide clinical variability. Glucose-6-phosphate dehydrogenase deficiency is the most common red cell enzyme disorder worldwide. Frequent clinical presentations include neonatal jaundice and episodic hemolysis after exposure to oxidative stress. Symptoms of pyruvate kinase deficiency and other glycolytic enzyme disorders include neonatal jaundice, chronic hemolytic anemia, gallstones, and transfusion-related and transfusion-independent iron overload. Diagnosis is critical for appropriate supportive care, monitoring, and treatment.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Glucosephosphate Dehydrogenase Deficiency/diagnosis , Glucosephosphate Dehydrogenase Deficiency/etiology , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/diagnosis , Pyruvate Metabolism, Inborn Errors/etiology , Glucosephosphate Dehydrogenase Deficiency/therapy , Humans , Pyruvate Kinase/therapeutic use
8.
Blood ; 131(20): 2183-2192, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29549173

ABSTRACT

An international, multicenter registry was established to collect retrospective and prospective clinical data on patients with pyruvate kinase (PK) deficiency, the most common glycolytic defect causing congenital nonspherocytic hemolytic anemia. Medical history and laboratory and radiologic data were retrospectively collected at enrollment for 254 patients with molecularly confirmed PK deficiency. Perinatal complications were common, including anemia that required transfusions, hyperbilirubinemia, hydrops, and prematurity. Nearly all newborns were treated with phototherapy (93%), and many were treated with exchange transfusions (46%). Children age 5 years and younger were often transfused until splenectomy. Splenectomy (150 [59%] of 254 patients) was associated with a median increase in hemoglobin of 1.6 g/dL and a decreased transfusion burden in 90% of patients. Predictors of a response to splenectomy included higher presplenectomy hemoglobin (P = .007), lower indirect bilirubin (P = .005), and missense PKLR mutations (P = .0017). Postsplenectomy thrombosis was reported in 11% of patients. The most frequent complications included iron overload (48%) and gallstones (45%), but other complications such as aplastic crises, osteopenia/bone fragility, extramedullary hematopoiesis, postsplenectomy sepsis, pulmonary hypertension, and leg ulcers were not uncommon. Overall, 87 (34%) of 254 patients had both a splenectomy and cholecystectomy. In those who had a splenectomy without simultaneous cholecystectomy, 48% later required a cholecystectomy. Although the risk of complications increases with severity of anemia and a genotype-phenotype relationship was observed, complications were common in all patients with PK deficiency. Diagnostic testing for PK deficiency should be considered in patients with apparent congenital hemolytic anemia and close monitoring for iron overload, gallstones, and other complications is needed regardless of baseline hemoglobin. This trial was registered at www.clinicaltrials.gov as #NCT02053480.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Genetic Association Studies , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/diagnosis , Adolescent , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Anemia, Hemolytic, Congenital Nonspherocytic/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Blood Transfusion , Child , Child, Preschool , Cholecystectomy/adverse effects , Cholecystectomy/methods , Combined Modality Therapy , Enzyme Activation , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation , Phenotype , Pyruvate Kinase/metabolism , Pyruvate Metabolism, Inborn Errors/etiology , Pyruvate Metabolism, Inborn Errors/metabolism , Pyruvate Metabolism, Inborn Errors/therapy , Splenectomy/adverse effects , Splenectomy/methods , Symptom Assessment , Treatment Outcome , Young Adult
9.
Int J Lab Hematol ; 39(2): 223-231, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28133914

ABSTRACT

INTRODUCTION: Pyruvate kinase (PK) deficiency is one of the most common hereditary nonspherocytic hemolytic anemias worldwide with clinical manifestations ranging from mild to severe hemolysis. However, investigation of this enzymopathy is lacking in Tunisia. We report here a pioneer investigation of PK deficiency among Tunisian cases referred to our laboratory for biological analysis of unknown cause of hemolytic anemia. METHODS: Two hundred and fifty-three patients with unknown cause of hemolytic anemia have been addressed to our laboratory in order to investigate for red blood cells genetic disorders. Red cell enzyme activities were measured by standard methods, and molecular analysis was performed by DNA sequencing. The interpretation of mutation effect and the molecular modeling were performed by using specific software. RESULTS: Six different PKLR mutations were found (c.966-1G>T; c.965+1G>A; c.721G>T; c.1163C>A; c.1456C>T; c.1537T>A), among which four are described for the first time. Genotype-phenotype correlations for the novel missense mutations were investigated by three-dimensional structure analysis. CONCLUSION: This study provides important data of PK deficiency among Tunisians. It might be followed by a large neonatal screening to determine the spectrum of PK mutations and identify potential deficient patients for an early medical follow-up.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Mutation, Missense/genetics , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/genetics , Anemia, Hemolytic/etiology , Anemia, Hemolytic/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , DNA Mutational Analysis , Erythrocytes/enzymology , Genetic Association Studies , Humans , Models, Molecular , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/etiology , Tunisia/epidemiology
10.
Int J Lab Hematol ; 36(3): 388-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24750686

ABSTRACT

Hereditary red blood cell enzymopathies are genetic disorders affecting genes encoding red blood cell enzymes. They cause a specific type of anemia designated hereditary nonspherocytic hemolytic anemia (HNSHA). Enzymopathies affect cellular metabolism, which, in the red cell, mainly consists of anaerobic glycolysis, the hexose monophosphate shunt, glutathione metabolism, and nucleotide metabolism. Enzymopathies are commonly associated with normocytic normochromic hemolytic anemia. In contrast to other hereditary red cell disorders such as membrane disorders or hemoglobinopathies, the morphology of the red blood cell shows no specific abnormalities. Diagnosis is based on detection of reduced specific enzyme activity and molecular characterization of the defect on the DNA level. The most common enzyme disorders are deficiencies of glucose-6-phosphate dehydrogenase (G6PD) and pyruvate kinase (PK). However, there are a number of other enzyme disorders, often much less known, causing HNSHA. These disorders are rare and often underdiagnosed, and the purpose of this review. In this brief review, we provide an overview of clinically relevant enzymes, their function in red cell metabolism, and key aspects of laboratory diagnosis.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Erythrocytes/enzymology , Erythrocytes/metabolism , Erythrocytes/pathology , Glycolysis , Hematologic Tests/methods , Humans
11.
Biochim Biophys Acta ; 1812(11): 1393-402, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21787864

ABSTRACT

Inherited glucose-6-phosphate isomerase (GPI) deficiency is the second most frequent glycolytic erythroenzymopathy in humans. Patients present with non-spherocytic anemia of variable severity and with neuromuscular dysfunction. We previously described Chinese hamster (CHO) cell lines with mutations in GPI and loss of GPI activity. This resulted in a temperature sensitivity and severe reduction in the synthesis of glycerolipids due to a reduction in phosphatidate phosphatase (PAP). In the current article we attempt to describe the nature of this pleiotropic effect. We cloned and sequenced the CHO lipin 1 cDNA, a gene that codes for PAP activity. Overexpression of lipin 1 in the GPI-deficient cell line, GroD1 resulted in increased PAP activity, however it failed to restore glycerolipid biosynthesis. Fluorescence microscopy showed a failure of GPI-deficient cells to localize lipin 1α to the nucleus. We also found that glucose-6-phosphate levels in GroD1 cells were 10-fold over normal. Lowering glucose levels in the growth medium partially restored glycerolipid biosynthesis and nuclear localization of lipin 1α. Western blot analysis of the elements within the mTOR pathway, which influences lipin 1 activity, was consistent with an abnormal activation of this system. Combined, these data suggest that GPI deficiency results in an accumulation of glucose-6-phosphate, and possibly other glucose-derived metabolites, leading to activation of mTOR and sequestration of lipin 1 to the cytosol, preventing its proper functioning. These results shed light on the mechanism underlying the pathologies associated with inherited GPI deficiency and the variability in the severity of the symptoms observed in these patients.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Cell Nucleus/metabolism , Glucose-6-Phosphate Isomerase/metabolism , Glucose/pharmacology , TOR Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Blotting, Western , CHO Cells , Cloning, Molecular , Cricetinae , Electrophoretic Mobility Shift Assay , Fructosephosphates/metabolism , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glycolipids , Microscopy, Fluorescence , Molecular Sequence Data , Organic Chemicals/metabolism , Phosphatidate Phosphatase/metabolism , Protein Transport , Sequence Homology, Amino Acid
12.
Haematologica ; 92(6): 731-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17550844

ABSTRACT

BACKGROUND AND OBJECTIVES: Red blood cell pyruvate kinase (R-PK) deficiency is the most common glycolytic enzyme defect associated with hereditary non-spherocytic hemolytic anemia. Cases with the most severe deficiency die in the peri- or neonatal period and no specific therapy exists at present. To test whether the targeted overexpression of the normal R-PK gene in erythroid cells could reduce hemolysis in R-PK mutant mice, we performed a genetic rescue study using human R-PK transgenic mice. DESIGN AND METHODS: Human R-PK promoter driven with human mLCR of the human b-globin locus was used for the erythroid-specific expression of human R-PK in murine erythrocytes. The transgenic lines were mated with homozygous R-PK mutant mice and subsequently backcrossed. Mutant homozygotes with the mLCR-R-PK transgene were examined for any therapeutic effects of transgene expression. RESULTS: Two PK transgenic lines, hRPK_lo and hRPK_hi, were obtained. R-PK activity of the transgenic mice reached as high as three times that of the animals with the endogenous PK gene. Overexpression of human R-PK in the homozygous mutant mice successfully reduced hemolytic anemia. Improvements of hemolysis were evaluated by hemoglobin concentration, reticulocyte count, and spleen weight, which showed significant correlations with the levels of expression of the transgene. Recovery from metabolic disturbance in mutant red blood cells was shown as normalized concentrations of the glycolytic intermediates upstream of PK. In addition, there was a remarkable negative correlation between R-PK activity and the number of TUNEL-positive erythroid progenitors in the spleen. INTERPRETATION AND CONCLUSIONS: These results indicate that overexpression of the wild-type PK gene in mutant erythroid cells ameliorates both erythroid apoptosis and the shortened red blood cell lifespan observed in PK mutant mice. It is likely that the level of transgene expression required to achieve evident therapeutic effects should be equivalent to or more than that of the endogenous PK gene. This gene-addition strategy may be suitable for clinical application if there is a high level of transgene expression of R-PK in erythroid progenitors/red blood cells.


Subject(s)
Anemia, Hemolytic/therapy , Genetic Therapy/methods , Pyruvate Kinase/deficiency , Anemia, Hemolytic/etiology , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Animals , Apoptosis , Erythrocyte Aging , Erythrocytes/enzymology , Erythrocytes/pathology , Humans , Mice , Mice, Transgenic , Pyruvate Kinase/genetics
15.
Blood ; 89(12): 4624-7, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9192788

ABSTRACT

We analyzed the molecular mutations of eight known Japanese glucose-6-phosphate dehydrogenase (G6PD) variants with unique biochemical properties. Three of them were caused by novel missense mutations: G6PD Musashino by 185 C-->T, G6PD Asahikawa by 695 G-->A, and G6PD Kamiube by 1387 C-->T. Predicted amino acid substitutions causing asymptomatic variants G6PD Musashino (62 Pro-->Phe) and G6PD Kamiube (463 Arg-->Cys) were located in regions near the amino or carboxyl end of the polypeptide chain, whereas an amino acid change 232 Cys-->Tyr causing a class 1 variant G6PD Asahikawa was located in the region where amino acid alterations in some class 1 variants were clustered. The other five variants had known missense mutations, namely, G6PD Fukushima, 1246 G-->A, G6PD Morioka, 1339 G-->A, and G6PD Iwate, G6PD Niigata and G6PD Yamaguchi, 1160 G-->A, which cause variants, G6PD Tokyo, G6PD Santiago de Cuba, and G6PD Beverly Hills, respectively.


Subject(s)
Asian People/genetics , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase/genetics , Point Mutation , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , DNA Mutational Analysis , Genetic Variation , Glucosephosphate Dehydrogenase/chemistry , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase Deficiency/complications , Glucosephosphate Dehydrogenase Deficiency/ethnology , Humans , Japan/epidemiology , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Structure-Activity Relationship
17.
Dtsch Med Wochenschr ; 118(36): 1276-80, 1993 Sep 10.
Article in German | MEDLINE | ID: mdl-8375297

ABSTRACT

An 11-year-old girl had marked haemolytic anaemia since the first year of life. Physical examination revealed scleral and cutaneous icterus and slight splenomegaly. Haemoglobin concentration was reduced to 9.5 g/dl, while platelet count and bilirubin concentration were increased (350,000/microliter and 2.2 mg/dl, respectively). The erythrocytes showed marked basophilic stippling, its extent typical of pyrimidine-5'-nucleotidase deficiency. The enzyme activity in the erythrocytes was 15% of normal. Deficiency of this enzyme, inherited as an autosomal recessive, is probably one of the most common erythrocyte enzyme deficiencies causing haemolytic anaemia. It brings about the intracellular accumulation of pyrimidine nucleotides which via secondary metabolic changes causes an accelerated destruction of erythrocytes. There is no known causative treatment: splenectomy is ineffective against the anaemia.


Subject(s)
5'-Nucleotidase/deficiency , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , 5'-Nucleotidase/blood , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Child , Consanguinity , Erythrocytes/enzymology , Female , Humans , Pedigree
18.
Blood ; 81(1): 206-13, 1993 Jan 01.
Article in English | MEDLINE | ID: mdl-8417789

ABSTRACT

The first two mutations causing hereditary glucose-6-phosphate isomerase (GPI) deficiency associated with chronic nonspherocytic hemolytic anemia in nonhuman mammals are described in the mouse. As in humans, the hemolytic syndrome, which is characterized by a diminished erythrocyte number, lower hematocrit, lower hemoglobin, higher number of reticulocytes and plasma bilirubin concentration, as well as increased liver- and spleen-somatic indices, was exclusively manifested in homozygous mutants. In comparison with wild type, heterozygous individuals exhibited neither hematologic differences nor alterations of other physiologic parameters, including plasma concentration of glucose, pyruvate and lactate, body weight, organo-somatic indices of liver, lung, kidney, spleen, and heart, as well as viability. Glycolytic intermediates, adenine nucleotides, and metabolic rate were not significantly altered in erythrocytes from heterozygotes. On the contrary, if allowance is made for the young erythrocyte population, homozygous mutant erythrocytes showed an increased concentration of glucose-6-phosphate and normal or decreased concentrations of glycolytic metabolites following the enzymatic block. The concentration of adenosine triphosphate and the glycolytic rate also appeared to be reduced. Homozygous anemic mice showed hepatosplenomegaly and typical adaptations to hypoxia, such as an elevated heart-somatic index and, for one mutant line, an enhanced lung-somatic index. Further, these animals were characterized by a marked reduction of body weight and an increase of lethality both correlated with the degree of enzyme deficiency in tissues. The latter findings were attributed to a reduced glycolytic capability of the whole organism caused by the enzyme defect in tissues, rather than representing secondary consequences of GPI deficiency in erythrocytes. The similarity in physicochemical and kinetic properties of the mutant murine proteins reported earlier with those of allozymes found in human GPI deficiency, as well as the comparable metabolic and physiologic consequences of this enzyme defect in mice and humans support that these murine mutants are excellent animal models for the human disease.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Disease Models, Animal , Adenine Nucleotides/blood , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Animals , Bilirubin/blood , Erythrocyte Count , Erythrocytes/metabolism , Glucose-6-Phosphate Isomerase/blood , Glucose-6-Phosphate Isomerase/genetics , Glycogen/metabolism , Glycolysis , Hematocrit , Heterozygote , Homozygote , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mutation , Organ Size , Reticulocytes/pathology , Spleen/pathology
20.
Acta Haematol Pol ; 23(2): 123-8, 1992.
Article in Polish | MEDLINE | ID: mdl-1488862

ABSTRACT

A therapeutic trial with mannose given intravenously as a 5% solution during 7 consecutive days (daily doses 12.5 g, 25 g and 50 g) was performed in a GPI deficient girl with nonspherocytic hemolytic anemia. The aim of the trial was to substitute glucose--the main red cells metabolic substrate--with mannose, since the glucose metabolism, due to the GPI deficiency, was significantly decreased. An initial good effect of treatment was disturbed with viral infection. No complications due to treatment with mannose were observed.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Hemolytic, Congenital Nonspherocytic/diet therapy , Erythrocytes/enzymology , Mannose/administration & dosage , Metabolism, Inborn Errors/blood , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Child , Female , Hematocrit , Hemoglobins/analysis , Humans , Infusions, Intravenous , Metabolism, Inborn Errors/complications , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...