Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Pediatr Blood Cancer ; 68(9): e29148, 2021 09.
Article in English | MEDLINE | ID: mdl-34125488

ABSTRACT

BACKGROUND: Pyruvate kinase deficiency (PKD) is a rare, autosomal recessive red blood cell enzyme disorder, which leads to lifelong hemolytic anemia and associated complications from the disease and its management. METHODS: An international, multicenter registry enrolled 124 individuals younger than 18 years old with molecularly confirmed PKD from 29 centers. Retrospective and prospective clinical data were collected. RESULTS: There was a wide range in the age at diagnosis from 0 to 16 years. Presentation in the newborn period ranged from asymptomatic to neonatal jaundice to fulminant presentations of fetal distress, myocardial depression, and/or liver failure. Children <5 years old were significantly more likely to be transfused than children >12 to <18 years (53% vs. 14%, p = .0006), which correlated with the timing of splenectomy. Regular transfusions were most common in children with two severe PKLR variants. In regularly transfused children, the nadir hemoglobin goal varied considerably. Impact on quality of life was a common reason for treatment with regular blood transfusions and splenectomy. Splenectomy increased the hemoglobin and decreased transfusion burden in most children but was associated with infection or sepsis (12%) and thrombosis (1.3%) even during childhood. Complication rates were high, including iron overload (48%), perinatal complications (31%), and gallstones (20%). CONCLUSIONS: There is a high burden of disease in children with PKD, with wide practice variation in monitoring and treatment. Clinicians must recognize the spectrum of the manifestations of PKD for early diagnostic testing, close monitoring, and management to avoid serious complications in childhood.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors , Adolescent , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Child , Child, Preschool , Humans , Prospective Studies , Pyruvate Metabolism, Inborn Errors/diagnosis , Pyruvate Metabolism, Inborn Errors/genetics , Pyruvate Metabolism, Inborn Errors/therapy , Quality of Life , Retrospective Studies
2.
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
4.
Am J Hematol ; 95(5): 472-482, 2020 05.
Article in English | MEDLINE | ID: mdl-32043619

ABSTRACT

Pyruvate kinase (PK) deficiency is a rare recessive congenital hemolytic anemia caused by mutations in the PKLR gene. This study reports the molecular features of 257 patients enrolled in the PKD Natural History Study. Of the 127 different pathogenic variants detected, 84 were missense and 43 non-missense, including 20 stop-gain, 11 affecting splicing, five large deletions, four in-frame indels, and three promoter variants. Within the 177 unrelated patients, 35 were homozygous and 142 compound heterozygous (77 for two missense, 48 for one missense and one non-missense, and 17 for two non-missense variants); the two most frequent mutations were p.R510Q in 23% and p.R486W in 9% of mutated alleles. Fifty-five (21%) patients were found to have at least one previously unreported variant with 45 newly described mutations. Patients with two non-missense mutations had lower hemoglobin levels, higher numbers of lifetime transfusions, and higher rates of complications including iron overload, extramedullary hematopoiesis, and pulmonary hypertension. Rare severe complications, including lower extremity ulcerations and hepatic failure, were seen more frequently in patients with non-missense mutations or with missense mutations characterized by severe protein instability. The PKLR genotype did not correlate with the frequency of complications in utero or in the newborn period. With ICCs ranging from 0.4 to 0.61, about the same degree of clinical similarity exists within siblings as it does between siblings, in terms of hemoglobin, total bilirubin, splenectomy status, and cholecystectomy status. Pregnancy outcomes were similar across genotypes in PK deficient women. This report confirms the wide genetic heterogeneity of PK deficiency.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Genetic Association Studies/methods , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Pyruvate Kinase/genetics , Young Adult
5.
Blood Cells Mol Dis ; 76: 59-62, 2019 05.
Article in English | MEDLINE | ID: mdl-30777730

ABSTRACT

Serum ferritin reflects total body iron stores, thus a low serum ferritin is used as a parameter of iron deficiency. In healthy adults in Japan, urine ferritin levels were about 5% of serum ferritin levels, with a correlation coefficient of 0.79. It is not known whether a low urine ferritin could serve as a non-invasive screen for iron deficiency. If so, this might be useful for neonates and young children, avoiding phlebotomy to screen for iron deficiency. However, for urinary ferritin screening to be feasible, ferritin must be measurable in the urine and correlate with serum ferritin. Testing should also clarify whether the iron content of ferritin in serum and urine are similar. In this pilot feasibility study we measured ferritin in paired serum and urine samples of healthy adult males, healthy term neonates, growing preterm neonates, and children who had very high serum ferritin levels from liver disorders or iron overload. We detected ferritin in every urine sample, and found a correlation with paired serum ferritin (Spearman correlation coefficient 0.78 of log10-transformed values). These findings suggest merit in further studying urinary ferritin in select populations, as a potential non-invasive screen to assess iron stores.


Subject(s)
Ferritins/blood , Ferritins/urine , Mass Screening/methods , Adult , Anemia, Iron-Deficiency , Child , Humans , Infant, Newborn , Japan , Liver Diseases , Male , Pilot Projects
7.
Blood Cells Mol Dis ; 72: 10-13, 2018 09.
Article in English | MEDLINE | ID: mdl-29880417

ABSTRACT

Herein we report a case series of seven newborn infants, all apparently well at birth, who in the period since 2009 were cared for in the State of Utah with acute bilirubin encephalopathy (ABE). This report summarizes our attempts to define common features of these seven through a state-wide voluntary registry, as a step toward devising new means of preventing such cases in the future. In previous reports of ABE, many of the affected neonates had no clearly defined explanation for their progressive hyperbilirubinemia. Our efforts to identify clear explanations in all seven cases included next generation DNA sequencing, testing a panel of 28 genes involved in bilirubin production and metabolism. We found that hemolytic disease was a unifying feature of these seven; two had DAT (+) Anti-D or anti-c hemolysis, while five had confirmed mutations in genes involved in bilirubin production and or metabolism that were previously unrecognized in these families.


Subject(s)
Bilirubin/genetics , Kernicterus/genetics , Bilirubin/analysis , Hemolysis , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Kernicterus/etiology , Mutation , Registries , Utah
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.
Blood Cells Mol Dis ; 71: 63-66, 2018 07.
Article in English | MEDLINE | ID: mdl-29599085

ABSTRACT

The congenital dyserythropoietic anemias are a heterogeneous group of disorders characterized by anemia and ineffective erythropoiesis. Congenital dyserythropoietic anemia type I (CDA1) can present in utero with hydrops fetalis, but more often it presents in childhood or adulthood with moderate macrocytic anemia, jaundice, and progressive iron-overload. CDA1 is inherited in an autosomal recessive manner, with biallelic pathogenic variants in CDAN1 or C15orf41. This case report documents a severe fetal presentation of CDA1 where we identified two novel compound heterozygous mutations in CDAN1 and describes the associated pathologic findings and levels of iron-regulatory proteins hepcidin, erythroferrone, and GDF15.


Subject(s)
Anemia, Dyserythropoietic, Congenital/diagnosis , Anemia, Dyserythropoietic, Congenital/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Glycoproteins/genetics , Heterozygote , Mutation , Adult , Anemia, Dyserythropoietic, Congenital/blood , Biomarkers , Biopsy , Bone Marrow , Female , Humans , Infant, Newborn , Male , Nuclear Proteins
10.
Clin Pediatr (Phila) ; 57(1): 19-26, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28090778

ABSTRACT

Various mutations in the genes encoding alpha spectrin (SPTA1) or beta spectrin (SPTB) are known to cause erythrocyte membrane disorders, sometimes associated with severe neonatal jaundice and anemia. We used a next-generation sequencing panel to evaluate 3 unrelated neonates who had puzzling cases of nonimmune hemolytic jaundice. In each case, we identified novel mutations in either SPTA1 or SPTB. Correlating erythrocyte morphology, clinical course, and computational analysis, we submit that each of the 3 variants is a probable pathogenic cause of the hereditary hemolytic conditions in these patients. We hope other pediatric practitioners caring for neonates with what appears to be idiopathic severe neonatal hyperbilirubinemia will look for spectrin variants as a possible cause, because additional cases with these specific variants along with this clinical phenotype are needed to confirm our postulate that these 3 cases are indeed pathogenic mutations.


Subject(s)
Elliptocytosis, Hereditary/genetics , Jaundice, Neonatal/genetics , Mutation/genetics , Spectrin/genetics , Spherocytosis, Hereditary/complications , Adult , Elliptocytosis, Hereditary/complications , Female , Humans , Infant, Newborn , Jaundice, Neonatal/complications , Jaundice, Neonatal/therapy , Male , Phototherapy
11.
Blood Cells Mol Dis ; 65: 38-40, 2017 06.
Article in English | MEDLINE | ID: mdl-28460265

ABSTRACT

In an iron deficient child, oral iron repeatedly failed to improve the condition. Whole exome sequencing identified one previously reported plus two novel mutation in the TMPRSS6 gene, with no mutations in other iron-associated genes. We propose that these mutations result in a novel variety of iron-refractory iron deficiency anemia.


Subject(s)
Anemia, Iron-Deficiency/genetics , Membrane Proteins/genetics , Mutation , Serine Endopeptidases/genetics , Alleles , Amino Acid Substitution , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Biomarkers , Blood Cell Count , DNA Mutational Analysis , Erythrocyte Indices , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Infant , Whole Genome Sequencing
12.
Pediatr Blood Cancer ; 64(5)2017 05.
Article in English | MEDLINE | ID: mdl-27808451

ABSTRACT

Symptomatic ß-thalassemia is one of the globally most common inherited disorders. The initial clinical presentation is variable. Although common hematological analyses are typically sufficient to diagnose the disease, sometimes the diagnosis can be more challenging. We describe a series of patients with ß-thalassemia whose diagnosis was delayed, required bone marrow examination in one affected member of each family, and revealed ringed sideroblasts, highlighting the association of this morphological finding with these disorders. Thus, in the absence of characteristic congenital sideroblastic mutations or causes of acquired sideroblastic anemia, the presence of ringed sideroblasts should raise the suspicion of ß-thalassemia.


Subject(s)
Anemia, Sideroblastic/pathology , Bone Marrow Cells/pathology , Erythroblasts/pathology , beta-Thalassemia/pathology , Adolescent , Adult , Anemia, Sideroblastic/diagnosis , Bone Marrow Cells/cytology , Bone Marrow Examination , Child , Erythroblasts/cytology , Erythrocytes, Abnormal , Female , Hematologic Diseases/complications , Humans , Infant , Male , beta-Thalassemia/diagnosis
14.
Curr Opin Pediatr ; 29(1): 12-19, 2017 02.
Article in English | MEDLINE | ID: mdl-27861255

ABSTRACT

PURPOSE OF REVIEW: As in adults and older children, anemia in newborn infants can be the result of erythropoietic failure, hemorrhage, or hemolysis. When hemolysis is the prime consideration, it can be challenging for physicians caring for neonates to choose from the wide variety of available diagnostic tests. This review describes the authors' opinions regarding rational, consistent, and cost-effective means of making an exact diagnosis of a neonatal hemolytic condition. RECENT FINDINGS: Two recent advances in the diagnosis of neonatal nonimmune hemolytic disorders are highlighted in this review: introduction of flow cytometry-based Eosin-5-maleimide (EMA) uptake as a screening test to identify RBC membrane defects and next-generation sequencing (NGS)-based panels to uncover exact mutations causing hemolysis. SUMMARY: The availability of newer tools such as EMA and NGS to diagnose specific hemolytic conditions, which might otherwise remain unknown, enables neonatal practitioners not only to identify the exact cause of hemolysis but also to discover novel mutations that can be implicated in the cause of neonatal hemolytic processes.


Subject(s)
Anemia, Hemolytic, Congenital/diagnosis , Anemia, Hemolytic, Congenital/blood , Anemia, Hemolytic, Congenital/etiology , Biomarkers/blood , Flow Cytometry , Genetic Markers , Humans , Infant, Newborn , Mutation , Sequence Analysis, DNA/methods
15.
Br J Haematol ; 174(5): 806-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27292444

ABSTRACT

Hereditary haemolytic anaemias are genetically and phenotypically heterogeneous disorders characterized by increased red cell destruction, with consequences ranging from innocuous to severe life-threatening anaemia. Diagnostic laboratories endeavour to assist clinicians reach the exact patient diagnosis by using tests principally based on morphological and biochemical techniques. However, these routine studies may be inconclusive, particularly in newborn infants and when transfusions have recently been administered. Large numbers and size of the potentially involved genes also impose a practical challenge for molecular diagnosis using routine sequencing approaches. To overcome these diagnostic shortcomings, we have utilized next-generation sequencing to provide a high-throughput, highly sensitive assay. We developed a panel interrogating 28 genes encoding cytoskeletal proteins and enzymes with sequencing coverage of the coding regions, splice site junctions, deep intronic and regulatory regions. We then evaluated 19 samples, including infants with unexplained extreme hyperbilirubinaemia and patients with transfusion-dependent haemolytic anaemia. Where possible, inheritance patterns of pathogenic mutations were determined by sequencing of immediate relatives. We conclude that this next-generation sequencing panel could be a cost-effective approach to molecular diagnosis of hereditary haemolytic anaemia, especially when the family history is uninformative or when routine laboratory testing fails to identify the causative haemolytic process.


Subject(s)
Anemia, Hemolytic, Congenital/diagnosis , High-Throughput Nucleotide Sequencing/methods , Adolescent , Adult , Anemia, Hemolytic, Congenital/genetics , Child , Child, Preschool , Cytoskeletal Proteins/genetics , Enzymes/genetics , Gene Components/genetics , High-Throughput Nucleotide Sequencing/economics , Humans , Hyperbilirubinemia, Hereditary/diagnosis , Infant , Infant, Newborn , Molecular Diagnostic Techniques/economics , Molecular Diagnostic Techniques/methods , Mutation , Young Adult
16.
Am J Med Genet A ; 170(9): 2449-52, 2016 09.
Article in English | MEDLINE | ID: mdl-27354418

ABSTRACT

Siblings presented as neonates with severe jaundice and transfusion-dependent hemolytic anemia. Next-generation sequencing revealed both to have three heterozygous mutations in the gene encoding erythrocyte pyruvate kinase (PKLR), plus a heterozygous splice mutation in the beta-spectrin gene (SPTB). In addition, both have a different 5th mutation in a gene encoding other erythrocyte membrane proteins. The asymptomatic parents and all three asymptomatic siblings have different sets of these mutations. © 2016 Wiley Periodicals, Inc.


Subject(s)
Genetic Association Studies , Genotype , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Siblings , Biomarkers , Female , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Mutation , Pedigree , Phenotype , Polymorphism, Genetic
17.
Clin Perinatol ; 43(2): 233-40, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27235204

ABSTRACT

Hemolysis can be an important cause of hyperbilirubinemia in premature and term neonates. It can result from genetic abnormalities intrinsic to or factors exogenous to normal to red blood cells (RBCs). Hemolysis can lead to a relatively rapid increase in total serum/plasma bilirubin, hyperbilirubinemia that is somewhat slow to fall with phototherapy, or hyperbilirubinemia that is likely to rebound after phototherapy. Laboratory methods for diagnosing hemolysis are more difficult to apply, or less conclusive, in preterm infants. Transfusion of donor RBCs can present a bilirubin load that must be metabolized. Genetic causes can be identified by next-generation sequencing panels.


Subject(s)
Bilirubin/metabolism , Cell Death , Erythrocytes , Hemolysis , Hyperbilirubinemia, Neonatal/metabolism , Blood Transfusion , Humans , Hyperbilirubinemia, Neonatal/therapy , Infant, Newborn , Infant, Premature , Phototherapy
18.
Clin Perinatol ; 42(3): 515-27, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26250914

ABSTRACT

A shortened erythrocyte life span, because of hemolytic disorders, is a common cause of extreme neonatal hyperbilirubinemia. Clinical and laboratory examinations can frequently identify the underlying cause of extreme neonatal hyperbilirubinemia. In this article, several tests, techniques, and approaches have been reviewed, including red blood cell morphology assessment, end-tidal carbon monoxide quantification, eosin-5-maleimide flow cytometry, as well as next-generation DNA sequencing using neonatal jaundice panels.


Subject(s)
Hematologic Diseases/complications , Hyperbilirubinemia, Neonatal/etiology , Humans , Hyperbilirubinemia, Neonatal/diagnosis , Infant, Newborn , Risk Factors , Severity of Illness Index
19.
Pediatrics ; 135(6): 1107-14, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26009624

ABSTRACT

Newborn infants who have hereditary spherocytosis (HS) can develop anemia and hyperbilirubinemia. Bilirubin-induced neurologic dysfunction is less likely in these neonates if the diagnosis of HS is recognized and appropriate treatment provided. Among neonates listed in the USA Kernicterus Registry, HS was the third most common underlying hemolytic condition after glucose-6-phosphate dehydrogenase deficiency and ABO hemolytic disease. HS is the leading cause of direct antiglobulin test (direct Coombs) negative hemolytic anemia requiring erythrocyte transfusion in the first months of life. We anticipate that as physicians become more familiar with diagnosing HS in the newborn period, fewer neonates with HS will develop hazardous hyperbilirubinemia or present to emergency departments with unanticipated symptomatic anemia. We predict that early suspicion, prompt diagnosis and treatment, and anticipatory guidance will prevent adverse outcomes in neonates with HS. The purpose of this article was to review the neonatal presentation of HS and to provide practical and up-to-date means of diagnosing and treating HS in neonates.


Subject(s)
Ankyrins/deficiency , Spherocytosis, Hereditary/diagnosis , Spherocytosis, Hereditary/therapy , Decision Trees , Humans , Infant, Newborn , Pediatrics , Practice Guidelines as Topic
20.
Transfusion ; 55(1): 187-96, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25082082

ABSTRACT

BACKGROUND: A recent NHLBI conference concluded that platelet (PLT) transfusions of neonates must become more evidence based. One neonatal disorder for which transfusions are given is a poorly defined entity, the "thrombocytopenia of perinatal asphyxia." To expand the evidence base for this entity, we performed a multicentered, retrospective analysis of neonates with perinatal asphyxia. STUDY DESIGN AND METHODS: We analyzed records of term and late preterm neonates with perinatal asphyxia defined by a cord blood pH of not more than 6.99 and/or base deficit of at least 16 mmol/L. From these we identified neonates with at least two PLT counts of fewer than 150 × 10(9) /L in the first week of life and described the severity, nadir, and duration of the thrombocytopenia. RESULTS: Thrombocytopenia occurred in 31% (117/375) of neonates with asphyxia versus 5% of matched nonasphyxiated controls admitted to a neonatal intensive care unit (p < 0.0001). Twenty-one of the 117 asphyxiated neonates were excluded from the remaining analysis due to disseminated intravascular coagulation or extracorporeal membrane oxygenation. Nadir PLT counts of the remaining 96 were on Day 3 (75 × 10(9) /L; 90% confidence interval, 35.7 × 10(9) -128.6 × 10(9) /L) and normalized by Days 19 to 21. PLT counts after asphyxia roughly correlated inversely with elevated nucleated red blood cell count (NRBC) counts at birth. Thirty of the 96 received at least one PLT transfusion, all given prophylactically, none for bleeding. CONCLUSIONS: We maintain that the thrombocytopenia of perinatal asphyxia is an authentic entity. Its association with elevated NRBC counts suggests that hypoxia is involved in the pathogenesis. Because PLT counts are only moderately low, the condition is transient, and bleeding problems seem rare, we speculate that PLT transfusions should not be needed for most neonates with this condition.


Subject(s)
Asphyxia Neonatorum/blood , Infant, Newborn/blood , Infant, Premature, Diseases/epidemiology , Infant, Premature/blood , Thrombocytopenia/epidemiology , Abruptio Placentae , Asphyxia Neonatorum/therapy , Cesarean Section , Datasets as Topic/statistics & numerical data , Disseminated Intravascular Coagulation/epidemiology , Disseminated Intravascular Coagulation/etiology , Extracorporeal Membrane Oxygenation , Female , Fetal Blood/chemistry , Hospitals, Voluntary/statistics & numerical data , Humans , Hydrogen-Ion Concentration , Hypothermia, Induced , Hypoxia/blood , Hypoxia/etiology , Incidence , Infant, Premature, Diseases/blood , Infant, Premature, Diseases/etiology , Infant, Premature, Diseases/therapy , Male , Platelet Count , Platelet Transfusion/statistics & numerical data , Pregnancy , Retrospective Studies , Thrombocytopenia/etiology , Thrombocytopenia/therapy , Treatment Outcome , Unnecessary Procedures
SELECTION OF CITATIONS
SEARCH DETAIL
...