Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
bioRxiv ; 2023 May 30.
Article in English | MEDLINE | ID: mdl-37398413

ABSTRACT

Mutation in nucleophosmin (NPM1) causes relocalization of this normally nucleolar protein to the cytoplasm ( NPM1c+ ). Despite NPM1 mutation being the most common driver mutation in cytogenetically normal adult acute myeloid leukemia (AML), the mechanisms of NPM1c+-induced leukemogenesis remain unclear. Caspase-2 is a pro-apoptotic protein activated by NPM1 in the nucleolus. Here, we show that caspase-2 is also activated by NPM1c+ in the cytoplasm, and DNA damage-induced apoptosis is caspase-2-dependent in NPM1c+ AML but not in NPM1wt cells. Strikingly, in NPM1c+ cells, loss of caspase-2 results in profound cell cycle arrest, differentiation, and down-regulation of stem cell pathways that regulate pluripotency including impairment in the AKT/mTORC1 and Wnt signaling pathways. In contrast, there were minimal differences in proliferation, differentiation, or the transcriptional profile of NPM1wt cells with and without caspase-2. Together, these results show that caspase-2 is essential for proliferation and self-renewal of AML cells that have mutated NPM1. This study demonstrates that caspase-2 is a major effector of NPM1c+ function and may even be a druggable target to treat NPM1c+ AML and prevent relapse.

2.
Br J Haematol ; 201(2): 343-352, 2023 04.
Article in English | MEDLINE | ID: mdl-36602125

ABSTRACT

Ischaemic stroke is a common complication of sickle cell disease (SCD) and without intervention can affect 11% of children with SCD before the age of 20. Within the Trans-Omics for Precision Medicine (TOPMed), a genome-wide association study (GWAS) of ischaemic stroke was performed on 1333 individuals with SCD from Brazil (178 cases, 1155 controls). Via a novel Cox proportional-hazards analysis, we searched for variants associated with ischaemic stroke occurring at younger ages. Variants at genome-wide significance (p < 5 × 10-8 ) include two near genes previously linked to non-SCD early-onset stroke (<65 years): ADAMTS2 (rs147625068, p = 3.70 × 10-9 ) and CDK18 (rs12144136, p = 2.38 × 10-9 ). Meta-analysis, which included the independent SCD cohorts Walk-PHaSST and PUSH, exhibited consistent association for variants rs1209987 near gene TBC1D32 (p = 3.36 × 10-10 ), rs188599171 near CUX1 (p = 5.89 × 10-11 ), rs77900855 near BTG1 (p = 4.66 × 10-8 ), and rs141674494 near VPS13C (1.68 × 10-9 ). Findings from this study support a multivariant model of early ischaemic stroke risk and possibly a shared genetic architecture between SCD individuals and non-SCD individuals younger than 65 years.


Subject(s)
Anemia, Sickle Cell , Brain Ischemia , Ischemic Stroke , Stroke , Adolescent , Adult , Child , Humans , Middle Aged , Young Adult , ADAMTS Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , Brain Ischemia/genetics , Brazil/epidemiology , Genome-Wide Association Study , Stroke/genetics
3.
Transl Res ; 252: 34-44, 2023 02.
Article in English | MEDLINE | ID: mdl-36041706

ABSTRACT

Overactive inflammatory responses are central to the pathophysiology of many hemolytic conditions including sickle cell disease. Excessive hemolysis leads to elevated serum levels of heme due to saturation of heme scavenging mechanisms. Extracellular heme has been shown to activate the NLRP3 inflammasome, leading to activation of caspase-1 and release of pro-inflammatory cytokines IL-1ß and IL-18. Heme also activates the non-canonical inflammasome pathway, which may contribute to NLRP3 inflammasome formation and leads to pyroptosis, a type of inflammatory cell death. Some clinical studies indicate there is a benefit to blocking the NLRP3 inflammasome pathway in patients with sickle cell disease and other hemolytic conditions. However, a thorough understanding of the mechanisms of heme-induced inflammasome activation is needed to fully leverage this pathway for clinical benefit. This review will explore the mechanisms of heme-induced NLRP3 inflammasome activation and the role of this pathway in hemolytic conditions including sickle cell disease.


Subject(s)
Anemia, Sickle Cell , Inflammasomes , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Heme/metabolism , Hemolysis , Inflammation/metabolism , Anemia, Sickle Cell/complications , Interleukin-1beta
4.
Curr Protoc ; 2(10): e582, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36300922

ABSTRACT

Vascular organ-chip or vessel-chip technology has significantly impacted our ability to model microphysiological vasculature. These biomimetic platforms have garnered significant interest from scientists and pharmaceutical companies as drug screening models. However, these models still lack the inclusion of patient-specific vasculature in the form of patient-derived endothelial cells. Blood outgrowth endothelial cells are patient blood-derived endothelial progenitors that have gained interest from the vascular biology community as an autologous endothelial cell alternative and have also been incorporated with the vessel-chip model. Next-generation sequencing techniques like RNA sequencing can further unlock the potential of personalized vessel-chips in discerning patient-specific hallmarks of endothelial dysfunction. Here we present a detailed protocol for (1) isolating blood outgrowth endothelial cells from patient blood samples, (2) culturing them in microfluidic vessel-chips, (3) isolating and preparing RNA from individual vessel-chips for sequencing, and (4) performing differential gene expression and bioinformatics analyses of vascular dysfunction and endothelial activation pathways. This method focuses specifically on identification of pathways and genes involved in vascular homeostasis and pathology, but can easily be adapted for the requirements of other systems. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Isolation of blood outgrowth endothelial cells from patient blood Basic Protocol 2: Culture of blood outgrowth endothelial cells in microfluidic vessel-chips Basic Protocol 3: Isolation of RNA from autologous vessel-chips Basic Protocol 4: Differential gene expression and bioinformatics analyses of endothelial activation pathways.


Subject(s)
Endothelial Cells , Transcriptome , Humans , Microfluidics/methods , Sequence Analysis, RNA , RNA/genetics
5.
Bioeng Transl Med ; 6(3): e10211, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34589594

ABSTRACT

Sickle cell disease (SCD) is the most prevalent inherited blood disorder in the world. But the clinical manifestations of the disease are highly variable. In particular, it is currently difficult to predict the adverse outcomes within patients with SCD, such as, vasculopathy, thrombosis, and stroke. Therefore, for most effective and timely interventions, a predictive analytic strategy is desirable. In this study, we evaluate the endothelial and prothrombotic characteristics of blood outgrowth endothelial cells (BOECs) generated from blood samples of SCD patients with known differences in clinical severity of the disease. We present a method to evaluate patient-specific vaso-occlusive risk by combining novel RNA-seq and organ-on-chip approaches. Through differential gene expression (DGE) and pathway analysis we find that BOECs from SCD patients exhibit an activated state through cell adhesion molecule (CAM) and cytokine signaling pathways among many others. In agreement with clinical symptoms of patients, DGE analyses reveal that patient with severe SCD had a greater extent of endothelial activation compared to patient with milder symptoms. This difference is confirmed by performing qRT-PCR of endothelial adhesion markers like E-selectin, P-selectin, tissue factor, and Von Willebrand factor. Finally, the differential regulation of the proinflammatory phenotype is confirmed through platelet adhesion readouts in our BOEC vessel-chip. Taken together, we hypothesize that these easily blood-derived endothelial cells evaluated through RNA-seq and organ-on-chips may serve as a biotechnique to predict vaso-occlusive episodes in SCD patients and will ultimately allow better therapeutic interventions.

6.
J Immunol ; 206(8): 1878-1889, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33741688

ABSTRACT

Excessive release of heme from RBCs is a key pathophysiological feature of several disease states, including bacterial sepsis, malaria, and sickle cell disease. This hemolysis results in an increased level of free heme that has been implicated in the inflammatory activation of monocytes, macrophages, and the endothelium. In this study, we show that extracellular heme engages the human inflammatory caspases, caspase-1, caspase-4, and caspase-5, resulting in the release of IL-1ß. Heme-induced IL-1ß release was further increased in macrophages from patients with sickle cell disease. In human primary macrophages, heme activated caspase-1 in an inflammasome-dependent manner, but heme-induced activation of caspase-4 and caspase-5 was independent of canonical inflammasomes. Furthermore, we show that both caspase-4 and caspase-5 are essential for heme-induced IL-1ß release, whereas caspase-4 is the primary contributor to heme-induced cell death. Together, we have identified that extracellular heme is a damage-associated molecular pattern that can engage canonical and noncanonical inflammasome activation as a key mediator of inflammation in macrophages.


Subject(s)
Anemia, Sickle Cell/metabolism , Caspases, Initiator/metabolism , Caspases/metabolism , Erythrocytes/physiology , Inflammasomes/metabolism , Inflammation/metabolism , Macrophages/immunology , Alarmins/metabolism , Cell Death , Cells, Cultured , Heme/metabolism , Hemolysis , Humans , Interleukin-1beta/metabolism , Up-Regulation
7.
Blood Adv ; 4(18): 4347-4357, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32915977

ABSTRACT

RHD and RHCE genes encode Rh blood group antigens and exhibit extensive single-nucleotide polymorphisms and chromosome structural changes in patients with sickle cell disease (SCD). RH variation can drive loss of antigen epitopes or expression of new epitopes, predisposing patients with SCD to Rh alloimmunization. Serologic antigen typing is limited to common Rh antigens, necessitating a genetic approach to detect variant antigen expression. We developed a novel algorithm termed RHtyper for RH genotyping from existing whole-genome sequencing (WGS) data. RHtyper determined RH genotypes in an average of 3.4 and 3.3 minutes per sample for RHD and RHCE, respectively. In a validation cohort consisting of 57 patients with SCD, RHtyper achieved 100% accuracy for RHD and 98.2% accuracy for RHCE, when compared with genotypes obtained by RH BeadChip and targeted molecular assays and after verification by Sanger sequencing and independent next-generation sequencing assays. RHtyper was next applied to WGS data from an additional 827 patients with SCD. In the total cohort of 884 patients, RHtyper identified 38 RHD and 28 RHCE distinct alleles, including a novel RHD DAU allele, RHD* 602G, 733C, 744T 1136T. RHtyper provides comprehensive and high-throughput RH genotyping from WGS data, facilitating deconvolution of the extensive RH genetic variation among patients with SCD. We have implemented RHtyper as a cloud-based public access application in DNAnexus (https://platform.dnanexus.com/app/RHtyper), enabling clinicians and researchers to perform RH genotyping with next-generation sequencing data.


Subject(s)
Blood Group Antigens , Rh-Hr Blood-Group System , Algorithms , Alleles , Genotype , Humans , Rh-Hr Blood-Group System/genetics
8.
Am J Hematol ; 95(4): 395-400, 2020 04.
Article in English | MEDLINE | ID: mdl-31903617

ABSTRACT

Although the most common front-line therapies for immune thrombocytopenia (ITP) have been in use for decades, it is still not possible to predict an individual patient's clinical course and response to therapy. Patients are managed with a trial-and-error approach and often suffer side effects of therapies which could have been avoided if response prediction were possible. Corticosteroids are the most frequently used upfront therapy for adults and children with ITP. Our group performed whole exome sequencing on a cohort of pediatric ITP patients, and identified two missense single nucleotide variants (SNV) in Toll-like receptor 4 (TLR4). These coding variants in TLR4 had an increased frequency in Caucasian patients with poor response to upfront steroid therapy. Both TLR4 (D299G; rs4986790) and TLR4 (T399I; rs4986791) had a minor allele frequency (MAF) of 20.7% in those patients unresponsive to steroids, but were present at lower allele frequencies of 2.3% and 3.4% in responders respectively (P < .001). These findings were consistent with the trend identified in an independent cohort of pediatric ITP patients treated with corticosteroids who underwent direct genotyping for both SNVs. This study identified two candidate genetic variants in two unique cohorts of ITP patients which may contribute to steroid response and have prognostic implications for treatment response in ITP.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Drug Resistance/genetics , Mutation, Missense , Polymorphism, Single Nucleotide , Purpura, Thrombocytopenic, Idiopathic/genetics , Toll-Like Receptor 4/genetics , Adrenal Cortex Hormones/pharmacokinetics , Alleles , Child , Child, Preschool , Cohort Studies , Exons/genetics , Female , Gene Frequency , Humans , Male , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Toll-Like Receptor 4/physiology , White People/genetics , Exome Sequencing
9.
J Clin Med ; 8(11)2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31694285

ABSTRACT

Chronic blood transfusions in patients with sickle cell anemia (SCA) cause iron overload, which occurs with a degree of interpatient variability in serum ferritin and liver iron content (LIC). Reasons for this variability are unclear and may be influenced by genes that regulate iron metabolism. We evaluated the association of the copy number of the glutathione S-transferase M1 (GSTM1) gene and degree of iron overload among patients with SCA. We compared LIC in 38 children with SCA and ≥12 lifetime erythrocyte transfusions stratified by GSTM1 genotype. Baseline LIC was measured using magnetic resonance imaging (MRI), R2*MRI within 3 months prior to, and again after, starting iron unloading therapy. After controlling for weight-corrected transfusion burden (mL/kg) and splenectomy, mean pre-chelation LIC (mg/g dry liver dry weight) was similar in all groups: GSTM1 wild-type (WT) (11.45, SD±6.8), heterozygous (8.2, SD±4.52), and homozygous GSTM1 deletion (GSTM1-null; 7.8, SD±6.9, p = 0.09). However, after >12 months of chelation, GSTM1-null genotype subjects had the least decrease in LIC compared to non-null genotype subjects (mean LIC change for GSTM1-null = 0.1 (SD±3.3); versus -0.3 (SD±3.0) and -1.9 (SD±4.9) mg/g liver dry weight for heterozygous and WT, respectively, p = 0.047). GSTM1 homozygous deletion may prevent effective chelation in children with SCA and iron overload.

10.
Lab Chip ; 19(15): 2500-2511, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31246211

ABSTRACT

Development of therapeutic approaches to treat vascular dysfunction and thrombosis at disease- and patient-specific levels is an exciting proposed direction in biomedical research. However, this cannot be achieved with animal preclinical models alone, and new in vitro techniques, like human organ-on-chips, currently lack inclusion of easily obtainable and phenotypically-similar human cell sources. Therefore, there is an unmet need to identify sources of patient primary cells and apply them in organ-on-chips to increase personalized mechanistic understanding of diseases and to assess drugs. In this study, we provide a proof-of-feasibility of utilizing blood outgrowth endothelial cells (BOECs) as a disease-specific primary cell source to analyze vascular inflammation and thrombosis in vascular organ-chips or "vessel-chips". These blood-derived BOECs express several factors that confirm their endothelial identity. The vessel-chips are cultured with BOECs from healthy or diabetic patients and form an intact 3D endothelial lumen. Inflammation of the BOEC endothelium with exogenous cytokines reveals vascular dysfunction and thrombosis in vitro similar to in vivo observations. Interestingly, our study with vessel-chips also reveals that unstimulated BOECs of type 1 diabetic pigs show phenotypic behavior of the disease - high vascular dysfunction and thrombogenicity - when compared to control BOECs or normal primary endothelial cells. These results demonstrate the potential of organ-on-chips made from autologous endothelial cells obtained from blood in modeling vascular pathologies and therapeutic outcomes at a disease and patient-specific level.


Subject(s)
Blood Cells/pathology , Endothelial Progenitor Cells/pathology , Lab-On-A-Chip Devices , Thrombosis/pathology , Adult , Cell Proliferation , Feasibility Studies , Humans , Oxidative Stress , Thrombosis/blood
11.
Blood Adv ; 1(18): 1414-1422, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-29296782

ABSTRACT

RH genes are highly polymorphic and encode the most complex of the 35 human blood group systems. This genetic diversity contributes to Rh alloimmunization in patients with sickle cell anemia (SCA) and is not avoided by serologic Rh-matched red cell transfusions. Standard serologic testing does not distinguish variant Rh antigens. Single nucleotide polymorphism (SNP)-based DNA arrays detect many RHD and RHCE variants, but the number of alleles tested is limited. We explored a next-generation sequencing (NGS) approach using whole-exome sequencing (WES) in 27 Rh alloimmunized and 27 matched non-alloimmunized patients with SCA who received chronic red cell transfusions and were enrolled in a multicenter study. We demonstrate that WES provides a comprehensive RH genotype, identifies SNPs not interrogated by DNA array, and accurately determines RHD zygosity. Among this multicenter cohort, we demonstrate an association between an altered RH genotype and Rh alloimmunization: 52% of Rh immunized vs 19% of non-immunized patients expressed variant Rh without co-expression of the conventional protein. Our findings suggest that RH allele variation in patients with SCA is clinically relevant, and NGS technology can offer a comprehensive alternative to targeted SNP-based testing. This is particularly relevant as NGS data becomes more widely available and could provide the means for reducing Rh alloimmunization in children with SCA.

12.
Blood Cells Mol Dis ; 62: 32-37, 2016 11.
Article in English | MEDLINE | ID: mdl-27838552

ABSTRACT

High levels of fetal hemoglobin (HbF) reduce sickle cell anemia (SCA) morbidity and mortality. HbF levels vary considerably and there is a strong genetic component that influences HbF production. Genetic polymorphisms at three quantitative trait loci (QTL): Xmn1-HBG2, HMIP-2 and BCL11A, have been shown to influence HbF levels and disease severity in SCA. Hydroxyurea (HU) is a drug that increases HbF. We investigated the influence of single nucleotide polymorphisms (SNPs) at the Xmn1-HBG2 (rs7482144); BCL11A (rs1427407, rs4671393 and rs11886868); and HMIP-2 (rs9399137 and rs9402686) loci on baseline and HU-induced HbF levels in 111 HbSS patients. We found that both BCL11A and HMIP-2 were associated with increased endogenous levels of HbF. Interestingly, we also found that BCL11A was associated with higher induction of HbF with HU. This effect was independent of the effect of BCL11A on baseline HbF levels. Additional studies will be needed to validate these findings and explain the ample inter-individual variations in HbF levels at baseline and HU-induced in patients with SCA.


Subject(s)
Carrier Proteins/genetics , Fetal Hemoglobin/analysis , Hydroxyurea/pharmacology , Metalloendopeptidases/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide/physiology , Adolescent , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Brazil , Child , Child, Preschool , Female , Fetal Hemoglobin/drug effects , Humans , Male , Middle Aged , Repressor Proteins , Young Adult
13.
PLoS One ; 11(10): e0164364, 2016.
Article in English | MEDLINE | ID: mdl-27711207

ABSTRACT

Discovery and validation of genetic variants that influence disease severity in children with sickle cell anemia (SCA) could lead to early identification of high-risk patients, better screening strategies, and intervention with targeted and preventive therapy. We hypothesized that newly identified genetic risk factors for the general African American population could also impact laboratory biomarkers known to contribute to the clinical disease expression of SCA, including variants influencing the white blood cell count and the development of albuminuria and abnormal glomerular filtration rate. We first investigated candidate genetic polymorphisms in well-characterized SCA pediatric cohorts from three prospective NHLBI-supported clinical trials: HUSTLE, SWiTCH, and TWiTCH. We also performed whole exome sequencing to identify novel genetic variants, using both a discovery and a validation cohort. Among candidate genes, DARC rs2814778 polymorphism regulating Duffy antigen expression had a clear influence with significantly increased WBC and neutrophil counts, but did not affect the maximum tolerated dose of hydroxyurea therapy. The APOL1 G1 polymorphism, an identified risk factor for non-diabetic renal disease, was associated with albuminuria. Whole exome sequencing discovered several novel variants that maintained significance in the validation cohorts, including ZFHX4 polymorphisms affecting both the leukocyte and neutrophil counts, as well as AGGF1, CYP4B1, CUBN, TOR2A, PKD1L2, and CD163 variants affecting the glomerular filtration rate. The identification of robust, reliable, and reproducible genetic markers for disease severity in SCA remains elusive, but new genetic variants provide avenues for further validation and investigation.


Subject(s)
Albuminuria/diagnosis , Anemia, Sickle Cell/diagnosis , Adolescent , Albuminuria/complications , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Apolipoprotein L1 , Apolipoproteins/genetics , Child , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Female , Genetic Variation , Genotype , Glomerular Filtration Rate , Homeodomain Proteins/genetics , Humans , Hydroxyurea/therapeutic use , Leukocyte Count , Leukocytes/cytology , Lipoproteins, HDL/genetics , Male , Neutrophils/cytology , Phenotype , Polymorphism, Single Nucleotide , Prospective Studies , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Risk Factors , Sequence Analysis, DNA , Transcription Factors/genetics
14.
PLoS One ; 9(10): e110740, 2014.
Article in English | MEDLINE | ID: mdl-25360671

ABSTRACT

Hydroxyurea has proven efficacy in children and adults with sickle cell anemia (SCA), but with considerable inter-individual variability in the amount of fetal hemoglobin (HbF) produced. Sibling and twin studies indicate that some of that drug response variation is heritable. To test the hypothesis that genetic modifiers influence pharmacological induction of HbF, we investigated phenotype-genotype associations using whole exome sequencing of children with SCA treated prospectively with hydroxyurea to maximum tolerated dose (MTD). We analyzed 171 unrelated patients enrolled in two prospective clinical trials, all treated with dose escalation to MTD. We examined two MTD drug response phenotypes: HbF (final %HbF minus baseline %HbF), and final %HbF. Analyzing individual genetic variants, we identified multiple low frequency and common variants associated with HbF induction by hydroxyurea. A validation cohort of 130 pediatric sickle cell patients treated to MTD with hydroxyurea was genotyped for 13 non-synonymous variants with the strongest association with HbF response to hydroxyurea in the discovery cohort. A coding variant in Spalt-like transcription factor, or SALL2, was associated with higher final HbF in this second independent replication sample and SALL2 represents an outstanding novel candidate gene for further investigation. These findings may help focus future functional studies and provide new insights into the pharmacological HbF upregulation by hydroxyurea in patients with SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Exome/genetics , Fetal Hemoglobin/metabolism , Genomics , Hydroxyurea/pharmacology , Sequence Analysis, DNA , Anemia, Sickle Cell/metabolism , Child , Cohort Studies , DNA-Binding Proteins , Dose-Response Relationship, Drug , Exome/drug effects , Humans , Hydroxyurea/adverse effects , Hydroxyurea/therapeutic use , Maximum Tolerated Dose , Open Reading Frames/genetics , Phenotype , Prospective Studies , Transcription Factors/genetics , Treatment Outcome
15.
Am J Hematol ; 88(7): 571-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23606168

ABSTRACT

The recently completed BABY HUG trial investigated the safety and efficacy of hydroxyurea in infants with sickle cell anemia (SCA). To investigate the effects of known genetic modifiers, genomic DNA on 190 randomized subjects were analyzed for alpha thalassemia, beta-globin haplotype, polymorphisms affecting endogenous fetal hemoglobin (HbF) levels (XmnI, BCL11A, and HBS1L-MYB), UGT1A1 promoter polymorphisms, and the common G6PD A(-) mutation. At study entry, infants with alpha thalassemia trait had significantly lower mean corpuscular volume, total bilirubin, and absolute reticulocyte count. Beta-globin haplotypes associated with milder disease had significantly higher hemoglobin and %HbF. BCL11A and XmnI polymorphisms had significant effects on baseline HbF, while UGT1A1 promoter polymorphisms significantly influenced baseline serum bilirubin. At study exit, subjects randomized to placebo still exhibited laboratory effects of alpha thalassemia and other modifiers, while those assigned hydroxyurea had treatment effects that exceeded most genetic influences. The pain phenotype was influenced by HbF modifiers in both treatment groups. These data document that genetic polymorphisms do modify laboratory and clinical phenotypes even in very young patients with SCA. The hydroxyurea effects are more potent, however, indicating that treatment criteria should not be limited to certain genetic subsets, and supporting the use of hydroxyurea for all young patients with SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Antisickling Agents/therapeutic use , Hydroxyurea/therapeutic use , Polymorphism, Genetic , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Bilirubin/blood , Carrier Proteins/blood , Carrier Proteins/genetics , Child, Preschool , Deoxyribonucleases, Type II Site-Specific/genetics , Female , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Glucosephosphate Dehydrogenase/blood , Glucosephosphate Dehydrogenase/genetics , Glucuronosyltransferase/blood , Glucuronosyltransferase/genetics , Haplotypes , Humans , Infant , Male , Nuclear Proteins/blood , Nuclear Proteins/genetics , Oncogene Proteins v-myb/blood , Oncogene Proteins v-myb/genetics , Promoter Regions, Genetic , Repressor Proteins , alpha-Thalassemia , beta-Globins/genetics , beta-Globins/metabolism
16.
Blood ; 121(16): 3237-45, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23422753

ABSTRACT

Stroke is a devastating complication of sickle cell anemia (SCA), occurring in 11% of patients before age 20 years. Previous studies of sibling pairs have demonstrated a genetic component to the development of cerebrovascular disease in SCA, but few candidate genetic modifiers have been validated as having a substantial effect on stroke risk. We performed an unbiased whole-genome search for genetic modifiers of stroke risk in SCA. Genome-wide association studies were performed using genotype data from single-nucleotide polymorphism arrays, whereas a pooled DNA approach was used to perform whole-exome sequencing. In combination, 22 nonsynonymous variants were identified and represent key candidates for further in-depth study. To validate the association of these mutations with the risk for stroke, the 22 candidate variants were genotyped in an independent cohort of control patients (n = 231) and patients with stroke (n = 57) with SCA. One mutation in GOLGB1 (Y1212C) and another mutation in ENPP1 (K173Q) were confirmed as having significant associations with a decreased risk for stroke. These mutations were discovered and validated by an unbiased whole-genome approach, and future studies will focus on how these functional mutations may lead to protection from stroke in the context of SCA.


Subject(s)
Anemia, Sickle Cell/complications , Membrane Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Polymorphism, Single Nucleotide , Pyrophosphatases/genetics , Stroke/etiology , Stroke/genetics , Adolescent , Anemia, Sickle Cell/genetics , Child , Child, Preschool , Cohort Studies , Exome , Genome-Wide Association Study , Golgi Matrix Proteins , Humans , Mutation , Risk Factors
17.
Br J Haematol ; 157(2): 240-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22360576

ABSTRACT

Sickle cell anaemia (SCA) is a severe debilitating haematological disorder associated with a high degree of morbidity and mortality. The level of fetal haemoglobin (HbF) is well-recognized as a critical laboratory parameter: lower HbF is associated with a higher risk of vaso-occlusive complications, organ damage, and early death. Hydroxycarbamide treatment can induce HbF, improve laboratory parameters, and ameliorate clinical complications of SCA but its mechanisms of action remain incompletely defined and the HbF response is highly variable. To identify pathways of hydroxycarbamide activity, we performed microarray expression analyses of early reticulocyte RNA obtained from children with SCA enrolled in the HydroxyUrea Study of Long-term Effects (NCT00305175) and examined the effects of hydroxycarbamide exposure in vivo. Hydroxycarbamide affected a large number of erythroid genes, with significant decreases in the expression of genes involved in translation, ribosome assembly and chromosome organization, presumably reflecting the daily cytotoxic pulses of hydroxycarbamide. Hydroxycarbamide also affected expression of numerous genes associated with HbF including BCL11A, a key regulator of baseline HbF levels. Together, these data indicate that hydroxycarbamide treatment for SCA leads to substantial changes in erythroid gene expression, including BCL11A and other potential signalling pathways associated with HbF induction.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Antisickling Agents/adverse effects , Gene Expression Regulation/drug effects , Hydroxyurea/adverse effects , Adolescent , Antisickling Agents/administration & dosage , Carrier Proteins/biosynthesis , Child , Child, Preschool , Female , Fetal Hemoglobin/biosynthesis , Gene Expression Profiling , Humans , Hydroxyurea/administration & dosage , Infant , Male , Nuclear Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Repressor Proteins , Signal Transduction/drug effects
18.
Pediatr Blood Cancer ; 59(2): 254-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22012708

ABSTRACT

BACKGROUND: The laboratory and clinical benefits of hydroxyurea therapy for children with sickle cell anemia (SCA) are well recognized, but treatment in young patients is limited in part by concerns about long-term genotoxicity, and specifically possible carcinogenicity. PROCEDURE: The Pediatric Hydroxyurea Phase III Clinical Trial (BABY HUG) was a multicenter double-blinded placebo-controlled randomized clinical trial (NCT00006400) testing whether hydroxyurea could prevent chronic organ damage in very young patients with SCA. An important secondary objective was the measurement of acquired genotoxicity using three laboratory assays: chromosomal karyotype, illegitimate VDJ recombination events, and micronucleated reticulocyte formation. RESULTS: Our data indicate that hydroxyurea treatment was not associated with any significant increases in genotoxicity compared to placebo treatment. CONCLUSIONS: These data provide additional support to the safety profile of hydroxyurea for young patients with SCA, and suggest that genotoxicity in this patient population is low.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Antisickling Agents/therapeutic use , DNA Damage/drug effects , DNA Damage/genetics , Hydroxyurea/therapeutic use , Double-Blind Method , Follow-Up Studies , Humans , Infant , Prognosis , V(D)J Recombination
19.
Blood ; 118(18): 4985-91, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21876119

ABSTRACT

Hydroxyurea therapy has proven laboratory and clinical efficacies for children with sickle cell anemia (SCA). When administered at maximum tolerated dose (MTD), hydroxyurea increases fetal hemoglobin (HbF) to levels ranging from 10% to 40%. However, interpatient variability of percentage of HbF (%HbF) response is high, MTD itself is variable, and accurate predictors of hydroxyurea responses do not currently exist. HUSTLE (NCT00305175) was designed to provide first-dose pharmacokinetics (PK) data for children with SCA initiating hydroxyurea therapy, to investigate pharmacodynamics (PD) parameters, including HbF response and MTD after standardized dose escalation, and to evaluate pharmacogenetics influences on PK and PD parameters. For 87 children with first-dose PK studies, substantial interpatient variability was observed, plus a novel oral absorption phenotype (rapid or slow) that influenced serum hydroxyurea levels and total hydroxyurea exposure. PD responses in 174 subjects were robust and similar to previous cohorts; %HbF at MTD was best predicted by 5 variables, including baseline %HbF, whereas MTD was best predicted by 5 variables, including serum creatinine. Pharmacogenetics analysis showed single nucleotide polymorphisms influencing baseline %HbF, including 5 within BCL11A, but none influencing MTD %HbF or dose. Accurate prediction of hydroxyurea treatment responses for SCA remains a worthy but elusive goal.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Hydroxyurea/pharmacokinetics , Hydroxyurea/therapeutic use , Pharmacogenetics , Adolescent , Biomarkers, Pharmacological/analysis , Biomarkers, Pharmacological/metabolism , Child , Child, Preschool , DNA Mutational Analysis , Dose-Response Relationship, Drug , Female , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacology , Male , Maximum Tolerated Dose , Pharmacogenetics/methods , Polymorphism, Single Nucleotide
20.
Br J Haematol ; 154(1): 134-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21542824

ABSTRACT

Hydroxycarbamide (hydroxyurea) provides laboratory and clinical benefits for adults and children with sickle cell anaemia (SCA). Given its mechanism of action and prior reports of genotoxicity, concern exists regarding long-term toxicities and possible carcinogenicity. We performed cross-sectional analyses of chromosome stability using peripheral blood mononuclear cells (PBMC) from 51 children with SCA and 3-12 years of hydroxycarbamide exposure (mean age 13·2 ± 4·1 years), compared to 28 children before treatment (9·4 ± 4·7 years). Chromosome damage was less for children receiving hydroxycarbamide than untreated patients (0·8 ± 1·2 vs. 1·9 ± 1·5 breaks per 100 cells, P = 0·004). There were no differences in repairing chromosome breaks after in vitro radiation; PBMC from children taking hydroxycarbamide had equivalent 2 Gy-induced chromosome breaks compared to untreated patients (30·8 ± 16·1 vs. 31·7 ± 8·9 per 100 cells, P = not significant). Radiation plus hydroxycarbamide resulted in similar numbers of unrepaired breaks in cells from children on hydroxycarbamide compared to untreated patients (95·8 ± 44·2 vs. 76·1 ± 23·1 per 100 cells, P = 0·08), but no differences were noted with longer exposure (97·9 ± 42·8 breaks per 100 cells for 3-6 years of hydroxycarbamide exposure vs. 91·2 ± 48·4 for 9-12 years of exposure). These observations provide important safety data regarding long-term risks of hydroxycarbamide exposure for children with SCA, and suggest low in vivo mutagenicity and carcinogenicity.


Subject(s)
Anemia, Sickle Cell/genetics , Antisickling Agents/adverse effects , Chromosome Aberrations/chemically induced , Hydroxyurea/adverse effects , Adolescent , Adult , Anemia, Sickle Cell/drug therapy , Antisickling Agents/administration & dosage , Cells, Cultured , Child , Child, Preschool , Cross-Sectional Studies , DNA Damage , DNA Repair , Drug Administration Schedule , Humans , Hydroxyurea/administration & dosage , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...