Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Community Genet ; 14(4): 355-360, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37391652

ABSTRACT

The objective of this study was to review the prevalence and features of the beta thalassaemia trait in Jamaican populations. Screening of 221,306 newborns over the last 46 years has given an indication of the distribution and prevalence of beta thalassaemia genes, and screening of 16,612 senior school students in Manchester parish, central Jamaica, has provided their haematological features. The prevalence of the beta thalassaemia trait predicted from double heterozygotes was 0.8% of 100,000 babies in Kingston, 0.9% of 121,306 newborns in southwest Jamaica, and 0.9% of school students in Manchester. Mild beta+ thalassaemia variants (-88 C>T, -29 A>G, -90 C>T, polyA T>C) accounted for 75% of Kingston newborns, 76% of newborns in southwest Jamaica, and 89% of Manchester students. Severe beta+ thalassaemia variants were uncommon. Betao thalassaemia variants occurred in 43 patients and resulted from 11 different variants of which the IVSII-849 A>G accounted for 25 (58%) subjects. Red cell indices in IVSII-781 C>G did not differ significantly from HbAA, and this is probably a harmless polymorphism rather than a form of beta+ thalassaemia; the removal of 6 cases in school screening had a minimal effect on the frequency of the beta thalassaemia trait. Red cell indices in the beta+ and betao thalassaemia traits followed established patterns, although both were associated with increased HbF levels. The benign nature of beta+ thalassaemia genes in Jamaica means that cases of sickle cell-beta+ thalassaemia are likely to be overlooked, and important clinical questions such as the role of pneumococcal prophylaxis remain to be answered.

2.
Hematology ; 28(1): 2184120, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36867085

ABSTRACT

We present a family that carries the ß-hemoglobin variant Hb Santa Juana (HBB:c.326A>G, ß 108(G10) Asn>Ser), also known as Hb Serres, in three generations. All affected family members had an anomal hemoglobin fraction as detected by HPLC but normal blood count without evidence of anemia or hemolysis. Oxygen affinity (p50 (O2) = 31.9-40.4 mmHg) was decreased in all probands, compared to 24.9-28.1 mmHg in unaffected individuals. Clinical symptoms likely related to the hemoglobin variant were cyanosis during anaesthesia, while other complaints such as shortness of breath or dizziness were less clearly linked with the hemoglobin variant.


Subject(s)
Cyanosis , Hemoglobins , Humans , Chromatography, High Pressure Liquid , Dyspnea , Oxygen
3.
J Community Genet ; 13(2): 229-234, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35018573

ABSTRACT

Based in the parish of Manchester in central Jamaica, the Manchester Project offered free detection of haemoglobin genotype to senior classes in 15 secondary schools between 2008 and 2013. Restricting the database to 15,103 students aged 15.0-19.9 years provided an opportunity to examine the red cell characteristics of the different haemoglobin genotypes, including normal (HbAA) in 85.0%, the sickle cell trait (HbAS) in 9.7%, HbC trait (HbAC) in 3.5% and hereditary persistence of foetal haemoglobin (HbA-HPFH) in 0.4%. Compared to the normal HbAA phenotype, HbAS had significantly increased mean cell haemoglobin concentration (MCHC), red cell count (RBC), and red cell distribution width (RDW) and decreased mean cell volume (MCV) and mean cell haemoglobin (MCH), these differences being even more marked in HbAC. Compared to HbAA, the HbA-HPFH had significantly increased RDW, but there were no consistent differences in other red cell indices, and there were no significant differences in haematological indices between the two common deletion HPFH variants, HPFH-1 and HPFH-2. Although these changes are unlikely to be clinically significant, they contribute to an understanding of the haematological spectrum of the common haemoglobin genotypes in peoples of African origin.

4.
Ann Hematol ; 100(7): 1647-1665, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33712866

ABSTRACT

Secondary thrombocytosis is a frequent secondary finding in childhood infection and inflammation. Primary hereditary thrombocytosis may be caused by germline mutations within the genes encoding key regulators of thrombopoiesis, i.e., thrombopoietin (THPO) and its receptor c-MPL (MPL) or the receptor's effector kinase Januskinase2 (JAK2). Furthermore, somatic mutations in JAK2, MPL, and in the gene-encoding calreticulin (CALR) have been described to act as driver mutations within the so-called Philadelphia-negative myeloproliferative neoplasms (MPNs), namely essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF). Increasing knowledge on the molecular mechanisms and on the clinical complications of these diseases is reflected by the WHO diagnostic criteria and European LeukemiaNet (ELN) recommendations on the management of adult MPN. However, data on childhood thrombocytosis are rare, and no consensus guidelines for pediatric thrombocytosis exist. Current literature has highlighted differences in the epidemiology and molecular pathogenesis of childhood thrombocytosis as compared to adults. Furthermore, age-dependent complications and pharmacological specificities suggest that recommendations tailored to the pediatric population are necessary in clinical practice. Here we summarize literature on classification, diagnostics, and clinical management of childhood thrombocytosis.


Subject(s)
Thrombocytosis , Adolescent , Adult , Age of Onset , Algorithms , Anticoagulants/therapeutic use , Calreticulin/genetics , Child , Disease Management , Female , Germ-Line Mutation , Humans , Hydroxyurea/therapeutic use , Interferon-alpha/therapeutic use , Janus Kinase 2/genetics , Male , Myelodysplastic-Myeloproliferative Diseases/complications , Platelet Count , Quinazolines/therapeutic use , Receptors, Thrombopoietin/genetics , Severity of Illness Index , Thrombocythemia, Essential/classification , Thrombocythemia, Essential/genetics , Thrombocytosis/classification , Thrombocytosis/diagnosis , Thrombocytosis/etiology , Thrombocytosis/therapy , Thrombophilia/drug therapy , Thrombophilia/etiology
5.
Leukemia ; 31(10): 2048-2056, 2017 10.
Article in English | MEDLINE | ID: mdl-28196983

ABSTRACT

Recent developments in sequencing technologies led to the discovery of a novel form of genomic instability, termed chromothripsis. This catastrophic genomic event, involved in tumorigenesis, is characterized by tens to hundreds of simultaneously acquired locally clustered rearrangements on one chromosome. We hypothesized that leukemias developing in individuals with Ataxia Telangiectasia, who are born with two mutated copies of the ATM gene, an essential guardian of genome stability, would show a higher prevalence of chromothripsis due to the associated defect in DNA double-strand break repair. Using whole-genome sequencing, fluorescence in situ hybridization and RNA sequencing, we characterized the genomic landscape of Acute Lymphoblastic Leukemia (ALL) arising in patients with Ataxia Telangiectasia. We detected a high frequency of chromothriptic events in these tumors, specifically on acrocentric chromosomes, as compared with tumors from individuals with other types of DNA repair syndromes (27 cases total, 10 with Ataxia Telangiectasia). Our data suggest that the genomic landscape of Ataxia Telangiectasia ALL is clearly distinct from that of sporadic ALL. Mechanistically, short telomeres and compromised DNA damage response in cells of Ataxia Telangiectasia patients may be linked with frequent chromothripsis. Furthermore, we show that ATM loss is associated with increased chromothripsis prevalence in additional tumor entities.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , Ataxia Telangiectasia/genetics , Neoplasm Proteins/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Ataxia Telangiectasia/complications , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Child , Child, Preschool , Chromosomes, Human/ultrastructure , Chromothripsis , DNA Repair/genetics , DNA, Neoplasm/genetics , Female , Genome, Human , Genomic Instability , Humans , In Situ Hybridization, Fluorescence , Male , Mutation , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasms/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Neoplasm/genetics , Sequence Analysis, DNA , Sequence Analysis, RNA , Telomere Shortening/genetics , Transcriptome
6.
Blood Cancer J ; 7(2): e523, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28157215

ABSTRACT

In the search for genes that define critical steps of relapse in pediatric T-cell acute lymphoblastic leukemia (T-ALL) and can serve as prognostic markers, we performed targeted sequencing of 313 leukemia-related genes in 214 patients: 67 samples collected at the time of relapse and 147 at initial diagnosis. As relapse-specific genetic events, we identified activating mutations in NT5C2 (P=0.0001, Fisher's exact test), inactivation of TP53 (P=0.0007, Fisher's exact test) and duplication of chr17:q11.2-24.3 (P=0.0068, Fisher's exact test) in 32/67 of T-ALL relapse samples. Alterations of TP53 were frequently homozygous events, which significantly correlated with higher rates of copy number alterations in other genes compared with wild-type TP53 (P=0.0004, Mann-Whitney's test). We subsequently focused on mutations with prognostic impact and identified genes governing DNA integrity (TP53, n=8; USP7, n=4; MSH6, n=4), having key roles in the RAS signaling pathway (KRAS, NRAS, n=8), as well as IL7R (n=4) and CNOT3 (n=4) to be exclusively mutated in fatal relapses. These markers recognize 24/49 patients with a second event. In 17 of these patients with mostly refractory relapse and dire need for efficient treatment, we identified candidate targets for personalized therapy with p53 reactivating compounds, MEK inhibitors or JAK/STAT-inhibitors that may be incorporated in future treatment strategies.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Child, Preschool , Disease-Free Survival , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Risk Factors
7.
Strahlenther Onkol ; 189(11): 957-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23801068

ABSTRACT

BACKGROUND: Osteosarcomas (OS) are highly malignant and radioresistant tumors. Histone deacetylase inhibitors (HDACi) constitute a novel class of anticancer agents. We sought to investigate the effect of combined treatment with suberoylanilide hydroxamic acid (SAHA) and radiotherapy in OS in vivo. METHODS: Clonogenic survival of human OS cell lines as well as tumor growth delay of OS xenografts were tested after treatment with either vehicle, radiotherapy (XRT), SAHA, or XRT and SAHA. Tumor proliferation, necrosis, microvascular density, apoptosis, and p53/p21 were monitored by immunohistochemistry. The CD95 pathway was performed by flow cytometry, caspase (3/7/8) activity measurements, and functional inhibition of CD95 death signaling. RESULTS: Combined treatment with SAHA and XRT markedly reduced the surviving fraction of OS cells as compared to XRT alone. Likewise, dual therapy significantly inhibited OS tumor growth in vivo as compared to XRT alone, reflected by reduced tumor proliferation, impaired angiogenesis, and increased apoptosis. Addition of HDACi to XRT led to elevated p53, p21, CD95, and CD95L expression. Inhibition of CD95 signaling reduced HDACi- and XRT-induced apoptosis. CONCLUSION: Our data show that HDACi increases the radiosensitivity of osteosarcoma cells at least in part via ligand-induced apoptosis. HDACi thus emerge as potentially useful treatment components of OS.


Subject(s)
Bone Neoplasms/physiopathology , Bone Neoplasms/therapy , Chemoradiotherapy/methods , Histone Deacetylase Inhibitors/therapeutic use , Osteosarcoma/physiopathology , Osteosarcoma/therapy , Radiation Tolerance/drug effects , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Humans , Mice , Mice, SCID , Osteosarcoma/pathology , Radiotherapy Dosage , Radiotherapy, Conformal/methods , Treatment Outcome
8.
Strahlenther Onkol ; 188(2): 168-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22249335

ABSTRACT

PURPOSE: Osteosarcoma and atypical teratoid rhabdoid tumors are tumor entities with varying response to common standard therapy protocols. Histone acetylation affects chromatin structure and gene expression which are considered to influence radiation sensitivity. The aim of this study was to investigate the effect of the combination therapy with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) and irradiation on atypical teratoid rhabdoid tumors and osteosarcoma compared to normal tissue cell lines. METHODS: Clonogenic assay was used to determine cell survival. DNA double-strand breaks (DSB) were examined by pulsed-field electrophoresis (PFGE) as well as by γH2AX immunostaining involving flow cytometry, fluorescence microscopy, and immunoblot analysis. RESULTS: SAHA lead to an increased radiosensitivity in tumor but not in normal tissue cell lines. γH2AX expression as an indicator for DSB was significantly increased when SAHA was applied 24 h before irradiation to the sarcoma cell cultures. In contrast, γH2AX expression in the normal tissue cell lines was significantly reduced when irradiation was combined with SAHA. Analysis of initial DNA fragmentation and fragment rejoining by PFGE, however, did not reveal differences in response to the SAHA pretreatment for either cell type. CONCLUSION: SAHA increases radiosensitivity in tumor but not normal tissue cell lines. The increased H2AX phosphorylation status of the SAHA-treated tumor cells post irradiation likely reflects its delayed dephosphorylation within the DNA damage signal decay rather than chromatin acetylation-dependent differences in the overall efficacy of DSB induction and rejoining. The results support the hypothesis that combining SAHA with irradiation may provide a promising strategy in the treatment of solid tumors.


Subject(s)
Histones/biosynthesis , Hydroxamic Acids/administration & dosage , Osteosarcoma/pathology , Osteosarcoma/radiotherapy , Radiation Tolerance/drug effects , Teratoma/pathology , Teratoma/radiotherapy , Cell Line, Tumor , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Histone Deacetylase Inhibitors/administration & dosage , Humans , Radiation Dosage , Radiation-Sensitizing Agents/administration & dosage , Treatment Outcome , Vorinostat
9.
Leukemia ; 24(12): 2005-13, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20944675

ABSTRACT

Precursor T-cell acute lymphoblastic leukemia (T-ALL) remains an important challenge in pediatric oncology. Because of the particularly poor prognosis of relapses, it is vital to identify molecular risk factors allowing early and effective treatment stratification. Activating NOTCH1 mutations signify a favorable prognosis in patients treated on ALL-BFM protocols. We have now tested if NOTCH pathway activation at different steps has similar clinical effects and if multiple mutations in this pathway function synergistically. Analysis of a validation set of 151 T-ALL patients and of the total cohort of 301 patients confirms the low relapse rate generally and the overall favorable effect of activating NOTCH1 mutations. Subgroup analysis shows that the NOTCH1 effect in ALL-BFM is restricted to patients with rapid early treatment response. Inactivation of the ubiquitin ligase FBXW7 is associated with rapid early treatment response and synergizes with NOTCH1 receptor activation. However, the effect of FBXW7 inactivation is separable from NOTCH1 activation by not synergizing with NOTCH1 mutations in predicting favorable long-term outcome, which can probably be explained by the interaction of FBXW7 with other clients. Finally, the comparison with other European protocols suggests that the NOTCH effect is treatment dependent generally and may depend on the intensity of central nervous system-directed therapy specifically.


Subject(s)
Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prednisone/therapeutic use , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/genetics , Cell Cycle Proteins/physiology , Child , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Humans , Treatment Outcome , Ubiquitin-Protein Ligases/physiology
10.
Klin Padiatr ; 218(3): 170-3, 2006.
Article in German | MEDLINE | ID: mdl-16688674

ABSTRACT

Homozygous sickle cell (HbSS) disease is paradigmatic for the complex influence of genetic modifiers of a monogenic disease. The genetically determined variability of HbF concentration has a strong impact on the clinical phenotype. The pharmacologically induced increase of HbF leads to a reduced morbidity which demonstrates that the knowledge of genetic modifiers enables the development of new therapeutic strategies. The presence of alpha-thalassemia also ameliorates the disease phenotype albeit not to the same extent as HbF does. Both factors, HbF and alpha-thalassemia are insufficient to explain the clinical variability of HbSS disease. The introduction of genome analysis has now provided the tools to identify relevant gene loci that will likely be helpful in estimating the probability of severe complications such as the occurrence of stroke. Following the validation in prospective studies, the subtle analysis of genetic modifiers will therefore influence the management of patients with sickle cell disease.


Subject(s)
Anemia, Sickle Cell/genetics , Genetic Therapy , Hemoglobin, Sickle/genetics , Homozygote , Anemia, Sickle Cell/mortality , Anemia, Sickle Cell/therapy , Child , Fetal Hemoglobin/genetics , Humans , Prognosis , Survival Rate , alpha-Thalassemia/genetics
11.
J Thromb Haemost ; 4(5): 1078-85, 2006 May.
Article in English | MEDLINE | ID: mdl-16689762

ABSTRACT

BACKGROUND: Mutations of the 3' end mRNA-processing signal of the prothrombin (F2) gene have been reported to cause elevated F2 plasma concentrations, thrombosis, and complications of pregnancy. Whereas the common F2 20210*A mutation is almost exclusively found in Caucasians, the F2 20209*T mutation has been reported in Afro-Americans and Afro-Caribbeans only. PATIENTS AND METHODS: Using LightCycler technology, three unrelated Jewish-Moroccan patients tested for obstetric complications were found to be carriers of the F2 20209*T allele. A detailed molecular analysis was performed to identify the functional impact of this mutation. RESULTS: We report three unrelated women of Jewish-Moroccan origin with a F2 20209*T mutation and fetal loss or infertility. The functional analysis revealed that the F2 20209*T mutation stimulates 3' end processing and up-regulates prothrombin protein expression as assessed by a highly sensitive luminescence-based reporter system. CONCLUSIONS: This is the first report of 20209*T in Caucasians, and functional analysis demonstrates that F2 20209*T falls into a general category of mutations of the F2 gene, which may possibly contribute to thrombophilia and complications of pregnancy by interfering with a tightly balanced architecture of non-canonical F2 3' end formation signals.


Subject(s)
Cytosine/chemistry , Jews , Mutation , Prothrombin/genetics , Thymine/chemistry , White People , Adult , Aged , Base Sequence , DNA Primers , Female , Humans , Male , Morocco/ethnology , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Klin Padiatr ; 216(6): 304-14, 2004.
Article in English | MEDLINE | ID: mdl-15565545

ABSTRACT

The exploration of the molecular origin of hereditary diseases focused on genes and proteins for many years. Recently, mRNA has gained increasing attention. Most human genes contain introns and a considerable proportion of transcripts are not only alternatively spliced but also regulated posttranscriptionally in manifold ways. mRNA processing as well as a complex network of interactions between the steps of gene expression and associated quality control mechanisms are guided by a multitude of regulative mRNA sequence elements. Therefore it is not surprising that mutations of such elements can cause or modify human disease. The purpose of this review is the illustration of principles of physiological and dysregulated mRNA metabolism as well as a survey of new analytical tools and therapeutic approaches.


Subject(s)
Genetic Diseases, Inborn/genetics , RNA, Messenger/metabolism , Base Sequence , Exons , Female , Gene Expression , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/therapy , Humans , Introns , Male , Microarray Analysis , Mutation , Point Mutation , Transcription, Genetic
14.
Acta Haematol ; 108(2): 102-5, 2002.
Article in English | MEDLINE | ID: mdl-12187030

ABSTRACT

Deficiency in glucose-6-phosphate dehydrogenase (G6PD) is the most common enzymopathy, and more than 125 different mutations causing G6PD deficiency have been identified. Chronic haemolytic anaemia (CHA) associated with G6PD deficiency is rare, but there is a cluster of mutations causing CHA between amino acids 361-428 which are encoded by exon 10 of the G6PD gene. This region is involved in the dimer formation of the active G6PD enzyme and therefore plays an important role for enzyme stability and activity. Here, we report a 17-year-old patient with CHA, who carries a rare G --> A mutation at nucleotide 1160 which causes an R387H amino acid substitution. We review the reports of the seven previously described patients with this mutation, concluding that G6PD deficiency should be considered as a rare differential diagnosis of chronic haemolytic, non-spherocytic anaemia.


Subject(s)
Anemia, Hemolytic, Congenital/genetics , Glucosephosphate Dehydrogenase Deficiency/complications , Adolescent , Amino Acid Substitution , Anemia, Hemolytic, Congenital/etiology , Chronic Disease , DNA Mutational Analysis , Diagnosis, Differential , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase Deficiency/diagnosis , Glucosephosphate Dehydrogenase Deficiency/genetics , Humans , Male , Point Mutation
15.
Nat Genet ; 28(4): 389-92, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11443298

ABSTRACT

The G-->A mutation at position 20210 of the prothrombin or coagulation factor II gene (F2) represents a common genetic risk factor for the occurrence of thromboembolic events. This mutation affects the 3'-terminal nucleotide of the 3' untranslated region (UTR) of the mRNA and causes elevated prothrombin plasma concentrations by an unknown mechanism. Here, we show that the mutation does not affect the amount of pre-mRNA, the site of 3' end cleavage or the length of the poly(A) tail of the mature mRNA. Rather, we demonstrate that the physiological F2 3' end cleavage signal is inefficient and that F2 20210 G-->A represents a gain-of-function mutation, causing increased cleavage site recognition, increased 3' end processing and increased mRNA accumulation and protein synthesis. Enhanced mRNA 3' end formation efficiency emerges as a novel principle causing a genetic disorder and explains the role of the F2 20210 G-->A mutation in the pathogenesis of thrombophilia. This work also illustrates the pathophysiologic importance of quantitatively minor aberrations of RNA metabolism.


Subject(s)
Prothrombin/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thrombophilia/genetics , 3' Untranslated Regions/genetics , 3' Untranslated Regions/metabolism , HeLa Cells , Humans , Immunoblotting , Prothrombin/biosynthesis , RNA Precursors/genetics , RNA Precursors/metabolism , Transcription, Genetic , Transfection
16.
EMBO J ; 20(3): 532-40, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157759

ABSTRACT

Premature translation termination codons are common causes of genetic disorders. mRNAs with such mutations are degraded by a surveillance mechanism termed nonsense-mediated decay (NMD), which represents a phylogenetically widely conserved post-transcriptional mechanism for the quality control of gene expression. How NMD-competent mRNPs are formed and specified remains a central question. Here, we have used human beta-globin mRNA as a model system to address the role of splicing and polyadenylation for human NMD. We show that (i) splicing is an indispensable component of the human beta-globin NMD pathway, which cannot be compensated for by exonic beta-globin 'failsafe' sequences; (ii) the spatial requirements of human beta-globin NMD, as signified by the maximal distance of the nonsense mutation to the final exon-exon junction, are less constrained than in yeast; and (iii) non-polyadenylated mRNAs with a histone 3' end are NMD competent. Thus, the formation of NMD-competent mRNP particles critically depends on splicing but does not require the presence of a poly(A) tail.


Subject(s)
Globins/genetics , RNA Splicing/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Base Sequence , Codon, Nonsense/genetics , DNA Primers/genetics , HeLa Cells , Humans , In Vitro Techniques , Mutation , Poly A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transfection
17.
Blood ; 96(10): 3357-63, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071627

ABSTRACT

The butyrate derivative isobutyramide (IBT) increases fetal hemoglobin (HbF) in patients with beta-hemoglobinopathies, but little is known about its usefulness for prolonged therapeutic use. We treated 8 patients with transfusion-dependent beta-thalassemia with 350 mg/kg of body weight per day of oral IBT for 126 to 384 days. During the trial period, the hemoglobin level was maintained between 85 g/L (range 82-87 g/L) (pretransfusion) and 115 g/L (range 110-119 g/L) (post-transfusion) (median, interquartile range), corresponding to 4-week transfusion intervals in all patients during the pretreatment phase. Adverse effects (bitter taste, epigastric discomfort) did not cause discontinuation of IBT. HbF increased in all patients from 3.1% (range 1.9%-4.8%) to 6.0% (range 3.3%-8.7) (P =.0017), while free Hb dropped from 0.48 g/L (range 0.39-0.81 g/L) to 0.19 g/L (range 0.16-0.24 g/L) (P <.0001). Transfusion intervals were consistently extended to 8 or 9 weeks in 1 patient, resulting in a decrease of daily iron load from 455 microgram/kg per day (range 451-459 microgram/kg per day) before therapy to 211 microgram/kg per day (range 203-286 microgram/kg per day) during the 12-month treatment period. Prolongation of transfusion intervals achieved by IBT was less consistent in another patient, whose parenteral iron load nevertheless decreased from 683 microgram/kg per day (range 618-748 microgram/kg per day) to 542 microgram/kg per day (340-596 microgram/kg per day). In the other 6 patients, no prolongation of transfusion intervals was achieved. Response to treatment was associated with high pretreatment HbF (> 4.5%), high parental HbF, and increased erythropoietin levels (> 150 IU/L). We conclude that IBT prolongs transfusion intervals and reduces parenteral iron burden in some patients with transfusion-dependent beta-thalassemia.


Subject(s)
Amides/pharmacology , Blood Transfusion , beta-Thalassemia/drug therapy , Administration, Oral , Adolescent , Adult , Amides/administration & dosage , Amides/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Blood/drug effects , Child , Child, Preschool , Drug Evaluation , Erythrocyte Indices/drug effects , Erythropoietin/blood , Female , Fetal Hemoglobin/drug effects , Fetal Hemoglobin/metabolism , Genotype , Hemoglobins/drug effects , Hemoglobins/metabolism , Hemolysis/drug effects , Homozygote , Humans , Iron/blood , Longitudinal Studies , Male , Patient Compliance , Time Factors , beta-Thalassemia/genetics , beta-Thalassemia/therapy
18.
Klin Padiatr ; 212(4): 159-62, 2000.
Article in English | MEDLINE | ID: mdl-10994543

ABSTRACT

BACKGROUND: Factor V Leiden (FVL) and Factor II (FII) G20210A represent common risk factors for thromboembolic (TE) events. In children, both venous and arterial TE-events have been associated with the presence of FVL and FII G20210A. In most heterozygous children with TE-events other prothrombotic factors can usually be identified. Case reports of children with homozygous FVL, including 3 patients described here, suggest that this genotype may convey a particulary high risk. However, prospective data about the type and frequency of TE-events in such children are lacking. STUDY DESIGN: We have initiated a prospective neonatal cohort study for the homozygous and double heterozygous genotypes for FVL and FII G20210A. The probands and the heterozygous controls are identified by neonatal screening that involves > 98% of the children born in Berlin and are followed up in a special out-patient clinic to document details of the clinical history, developmental parameters and the occurrence of TE-events. CONCLUSIONS: This study will provide controlled and unbiased information about the clinical significance of the homozygous and double heterozygous genotypes of these mutations.


Subject(s)
Blood Coagulation Disorders/complications , Factor V/genetics , Genetic Testing , Intracranial Thrombosis/diagnosis , Intracranial Thrombosis/genetics , Prothrombin/genetics , Blood Coagulation Disorders/genetics , Child , Child, Preschool , Female , Germany , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Intracranial Thrombosis/blood , Intracranial Thrombosis/complications , Male , Paresis/etiology , Prospective Studies , Research Design , Status Epilepticus/etiology
20.
Hum Mutat ; 15(3): 254-60, 2000.
Article in English | MEDLINE | ID: mdl-10679941

ABSTRACT

Phenylketonuria (PKU) is an important error of amino acid metabolism which results in most patients from phenylalanine hydroxylase (PAH) deficiency. PKU displays a marked genotypic heterogeneity both within and between different populations. The aim of this study was to establish the genotypic spectrum of PKU in eastern Germany, and to compare this to the distribution of mutations in western Germany. The study population included 302 patients in 290 families who were followed at treatment centers in Berlin, Leipzig and Jena. The study showed marked genotypic variability with a total of 75 mutations, including 15 that have so far not been described (eleven missense mutations, one splicing mutation, and three small deletions). One of these novel mutations, E183Q, occurred in cis to a R408W mutation. In the non-immigrant eastern German population, the frequency of R408W accounted for 40.1% of the PKU alleles. In the immigrant Turkish population of the former West Berlin, the most prevalent mutation was IVS10-11G>A (57%). There was a marked difference of the genotypic spectrum between the population studied here and the data reported from the western part of the country.


Subject(s)
Phenylalanine/blood , Phenylketonurias/genetics , Amino Acid Substitution , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Family Health , Female , Germany , Humans , Male , Mutation , Phenylketonurias/blood , Point Mutation , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...