1
|
Huang L, Zhang Q, Ye Y, Long Y, Huang H, Niu C, Lin B, Zeng L, Wang Y, Dai T, Hua X, Xu X. Rapid detection of genetic modifiers of β-thalassemia based on MALDI-TOF MS. Ann Hematol 2025; 104:1481-1492. [PMID: 40016399 PMCID: PMC12031963 DOI: 10.1007/s00277-025-06277-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
Fetal hemoglobin (HbF) levels are influenced by various genetic modifiers, which have clinically beneficial effects on both β-thalassemia and sickle cell disease. HbF-associated genetic variants are distributed throughout the genome, and current detection methods are often costly, time-consuming, and require multiple tests. Therefore, developing rapid and economical methods for the simultaneous detection of HbF-associated variants is essential for improving the accurate diagnosis of β-hemoglobinopathies. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) was employed to detect 20 well-documented genetic modifiers in BCL11A, KLF1, HBG2, DNMT1, GATAD2A, and HBS1L-MYB intergenic polymorphism (HMIP). The new assay's accuracy, repeatability, and lowest detection limit were evaluated. It was subsequently applied to 81 samples, and the clinical effects of the modifiers were further verified in a cohort of 560 β-thalassemia patients. The MALDI-TOF MS assays successfully detected all 20 genetic modifiers simultaneously in a single reaction. Genotyping results from 15 repetitions were consistent and accurate, indicating the stability of this assay. The assay's lowest detection limit for DNA was as low as 0.2 ng, sufficient for simultaneous genotyping of all loci. A double-blind evaluation of 81 samples showed 100% concordance with traditional genotyping methods. Significant differences were observed in HbF levels, survival time without transfusion, and clinical classification for the detected genetic modifiers. The MALDI-TOF MS detection assay for HbF-related variants is simple, rapid and high throughput. It enables the detection of 20 genetic modifiers in a single test, supporting accurate large-scale detection and enhancing the precise diagnosis and clinical classification of β-thalassemia.
Collapse
Affiliation(s)
- Li Huang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qianqian Zhang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, Guangdong, 523001, China
| | - Yuhua Ye
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yong Long
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Haoyang Huang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chao Niu
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Bin Lin
- Genetics Laboratory, Guangzhou Jiexu Gene Technology Co., Ltd, Guangzhou, Guangdong, 510530, China
| | - Lilan Zeng
- Genetics Laboratory, Guangzhou Jiexu Gene Technology Co., Ltd, Guangzhou, Guangdong, 510530, China
| | - Yuxi Wang
- Research and Development Center, Intelligene Biosystems (Qingdao) Co., Ltd, Qingdao, Shandong, 266114, China
| | - Tingting Dai
- Research and Development Center, Intelligene Biosystems (Qingdao) Co., Ltd, Qingdao, Shandong, 266114, China
| | - Xiaoyun Hua
- Genetics Laboratory, Guangzhou Jiexu Gene Technology Co., Ltd, Guangzhou, Guangdong, 510530, China.
| | - Xiangmin Xu
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
2
|
Musallam KM, Sheth S, Cappellini MD, Forni GL, Maggio A, Taher AT. Anemia and iron overload as prognostic markers of outcomes in β-thalassemia. Expert Rev Hematol 2024; 17:631-642. [PMID: 39037857 DOI: 10.1080/17474086.2024.2383420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Ineffective erythropoiesis and subsequent anemia as well as primary and secondary (transfusional) iron overload are key drivers for morbidity and mortality outcomes in patients with β-thalassemia. AREAS COVERED In this review, we highlight evidence from observational studies evaluating the association between measures of anemia and iron overload versus outcomes in both non-transfusion-dependent and transfusion-dependent forms of β-thalassemia. EXPERT OPINION Several prognostic thresholds have been identified with implications for patient management. These have also formed the basis for the design of novel therapy clinical trials by informing eligibility and target endpoints. Still, several data gaps persist in view of the challenge of assessing prospective long-term outcomes in a chronic disease. Pooling insights on the prognostic value of different measures of disease mechanism will be key to design future scoring systems that can help optimize patient management.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Sujit Sheth
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy
| | | | - Aurelio Maggio
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
3
|
Al-Allawi N, Atroshi SD, Sadullah RK, Eissa AA, Kriegshäuser G, Al-Zebari S, Qadir S, Khalil D, Oberkanins C. A Population-Oriented Genetic Scoring System to Predict Phenotype: A Pathway to Personalized Medicine in Iraqis With β-Thalassemia. Hemoglobin 2024:1-7. [PMID: 38390736 DOI: 10.1080/03630269.2024.2319733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
To assess the roles of genetic modifiers in Iraqi β-thalassemia patients, and determine whether a genotype-based scoring system could be used to predict phenotype, a total of 224 Iraqi patients with molecularly characterized homozygous or compound heterozygous β-thalassemia were further investigated for α-thalassemia deletions as well as five polymorphisms namely: rs7482144 C > T at HBG2, rs1427407 G > T and rs10189857 A > G at BCL11A, and rs28384513 A > C and rs9399137 T > C at HMIP. The enrolled patients had a median age of 14 years, with 96 males and 128 females. They included 144 thalassemia major, and 80 thalassemia intermedia patients. Multivariate logistic regression analysis revealed that a model including sex and four of these genetic modifiers, namely: β+ alleles, HBG2 rs7482144, α-thalassemia deletions, and BCL11A rs1427407 could significantly predict phenotype (major versus intermedia) with an overall accuracy of 83.9%. Furthermore, a log odds genetic score based on these significant predictors had a highly significant area under curve of 0.917 (95% CI 0.882-0.953). This study underscores the notion that genetic scoring systems should be tailored to populations in question, since genetic modifiers (and/or their relative weight) vary between populations. The population-oriented genetic scoring system created by the current study to predict β-thalassemia phenotype among Iraqis may pave the way to personalized medicine in this patient's group.
Collapse
Affiliation(s)
- Nasir Al-Allawi
- Department of Pathology, College of Medicine, University of Duhok, Iraq
| | - Sulav D Atroshi
- Department of Pathology, College of Medicine, University of Duhok, Iraq
| | - Regir K Sadullah
- Medical Laboratory Technology Department, College of Health and Medical Technology, Duhok Polytechnic University, Shekhan, Iraq
| | | | | | - Shaima Al-Zebari
- Research Center, College of Science, University of Duhok, Duhok, Iraq
| | - Shatha Qadir
- Department of Hematology, Azadi Teaching Hospital, Duhok, Iraq
| | - Dilan Khalil
- Research Center, College of Science, University of Duhok, Duhok, Iraq
| | - Christian Oberkanins
- Department of Research and Development, ViennaLab Diagnostics GmbH, Vienna, Austria
| |
Collapse
|
4
|
Gambari R, Zuccato C, Cosenza LC, Zurlo M, Gasparello J, Finotti A, Gamberini MR, Prosdocimi M. The Long Scientific Journey of Sirolimus (Rapamycin): From the Soil of Easter Island (Rapa Nui) to Applied Research and Clinical Trials on β-Thalassemia and Other Hemoglobinopathies. BIOLOGY 2023; 12:1202. [PMID: 37759601 PMCID: PMC10525103 DOI: 10.3390/biology12091202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
In this review article, we present the fascinating story of rapamycin (sirolimus), a drug able to induce γ-globin gene expression and increased production of fetal hemoglobin (HbF) in erythroid cells, including primary erythroid precursor cells (ErPCs) isolated from β-thalassemia patients. For this reason, rapamycin is considered of great interest for the treatment of β-thalassemia. In fact, high levels of HbF are known to be highly beneficial for β-thalassemia patients. The story of rapamycin discovery began in 1964, with METEI, the Medical Expedition to Easter Island (Rapa Nui). During this expedition, samples of the soil from different parts of the island were collected and, from this material, an antibiotic-producing microorganism (Streptomyces hygroscopicus) was identified. Rapamycin was extracted from the mycelium with organic solvents, isolated, and demonstrated to be very active as an anti-bacterial and anti-fungal agent. Later, rapamycin was demonstrated to inhibit the in vitro cell growth of tumor cell lines. More importantly, rapamycin was found to be an immunosuppressive agent applicable to prevent kidney rejection after transplantation. More recently, rapamycin was found to be a potent inducer of HbF both in vitro using ErPCs isolated from β-thalassemia patients, in vivo using experimental mice, and in patients treated with this compound. These studies were the basis for proposing clinical trials on β-thalassemia patients.
Collapse
Affiliation(s)
- Roberto Gambari
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (M.R.G.)
| | - Cristina Zuccato
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (M.R.G.)
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (L.C.C.); (M.Z.); (J.G.)
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (L.C.C.); (M.Z.); (J.G.)
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (L.C.C.); (M.Z.); (J.G.)
| | - Alessia Finotti
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (M.R.G.)
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (L.C.C.); (M.Z.); (J.G.)
| | - Maria Rita Gamberini
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (M.R.G.)
| | | |
Collapse
|
5
|
Finotti A, Gambari R. Combined approaches for increasing fetal hemoglobin (HbF) and de novo production of adult hemoglobin (HbA) in erythroid cells from β-thalassemia patients: treatment with HbF inducers and CRISPR-Cas9 based genome editing. Front Genome Ed 2023; 5:1204536. [PMID: 37529398 PMCID: PMC10387548 DOI: 10.3389/fgeed.2023.1204536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Genome editing (GE) is one of the most efficient and useful molecular approaches to correct the effects of gene mutations in hereditary monogenetic diseases, including β-thalassemia. CRISPR-Cas9 gene editing has been proposed for effective correction of the β-thalassemia mutation, obtaining high-level "de novo" production of adult hemoglobin (HbA). In addition to the correction of the primary gene mutations causing β-thalassemia, several reports demonstrate that gene editing can be employed to increase fetal hemoglobin (HbF), obtaining important clinical benefits in treated β-thalassemia patients. This important objective can be achieved through CRISPR-Cas9 disruption of genes encoding transcriptional repressors of γ-globin gene expression (such as BCL11A, SOX6, KLF-1) or their binding sites in the HBG promoter, mimicking non-deletional and deletional HPFH mutations. These two approaches (β-globin gene correction and genome editing of the genes encoding repressors of γ-globin gene transcription) can be, at least in theory, combined. However, since multiplex CRISPR-Cas9 gene editing is associated with documented evidence concerning possible genotoxicity, this review is focused on the possibility to combine pharmacologically-mediated HbF induction protocols with the "de novo" production of HbA using CRISPR-Cas9 gene editing.
Collapse
Affiliation(s)
- Alessia Finotti
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, University of Ferrara, Ferrara, Italy
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, University of Ferrara, Ferrara, Italy
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
6
|
The rs368698783 (G>A) Polymorphism Affecting LYAR Binding to the Aγ-Globin Gene Is Associated with High Fetal Hemoglobin (HbF) in β-Thalassemia Erythroid Precursor Cells Treated with HbF Inducers. Int J Mol Sci 2023; 24:ijms24010776. [PMID: 36614221 PMCID: PMC9821710 DOI: 10.3390/ijms24010776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
The human homologue of mouse Ly-1 antibody reactive clone protein (LYAR) is a putative novel regulator of γ-globin gene transcription. The LYAR DNA-binding motif (5′-GGTTAT-3′) is located within the 5′-UTR of the Aγ-globin gene. The LYAR rs368698783 (G>A) polymorphism is present in β-thalassemia patients and decreases the LYAR binding efficiency to the Aγ-globin gene. The objective of this study was to stratify β-thalassemia patients with respect to the rs368698783 (G>A) polymorphism and to verify whether their erythroid precursor cells (ErPCs) differentially respond in vitro to selected fetal hemoglobin (HbF) inducers. The rs368698783 (G>A) polymorphism was detected by DNA sequencing, hemoglobin production by HPLC, and accumulation of globin mRNAs by RT-qPCR. We found that the LYAR rs368698783 (G>A) polymorphism is associated with high basal and induced production of fetal hemoglobin in β-thalassemia patients. The most striking association was found using rapamycin as an HbF inducer. The results presented here could be considered important not only for basic biomedicine but also in applied translational research for precision medicine in personalized therapy of β-thalassemia. Accordingly, our data suggest that the rs368698783 polymorphism might be considered among the parameters useful to recruit patients with the highest probability of responding to in vivo hydroxyurea (HU) treatment.
Collapse
|
7
|
Zuccato C, Cosenza LC, Zurlo M, Gasparello J, Papi C, D’Aversa E, Breveglieri G, Lampronti I, Finotti A, Borgatti M, Scapoli C, Stievano A, Fortini M, Ramazzotti E, Marchetti N, Prosdocimi M, Gamberini MR, Gambari R. Expression of γ-globin genes in β-thalassemia patients treated with sirolimus: results from a pilot clinical trial (Sirthalaclin). Ther Adv Hematol 2022; 13:20406207221100648. [PMID: 35755297 PMCID: PMC9218916 DOI: 10.1177/20406207221100648] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/27/2022] [Indexed: 02/01/2023] Open
Abstract
Introduction β-thalassemia is caused by autosomal mutations in the β-globin gene, which induce the absence or low-level synthesis of β-globin in erythroid cells. It is widely accepted that a high production of fetal hemoglobin (HbF) is beneficial for patients with β-thalassemia. Sirolimus, also known as rapamycin, is a lipophilic macrolide isolated from a strain of Streptomyces hygroscopicus that serves as a strong HbF inducer in vitro and in vivo. In this study, we report biochemical, molecular, and clinical results of a sirolimus-based NCT03877809 clinical trial (a personalized medicine approach for β-thalassemia transfusion-dependent patients: testing sirolimus in a first pilot clinical trial, Sirthalaclin). Methods Accumulation of γ-globin mRNA was analyzed using reverse-transcription quantitative polymerase chain reaction (PCR), while the hemoglobin pattern was analyzed using high-performance liquid chromatography (HPLC). The immunophenotype was analyzed using a fluorescence-activated cell sorter (FACS), with antibodies against CD3, CD4, CD8, CD14, CD19, CD25 (for analysis of peripheral blood mononuclear cells), or CD71 and CD235a (for analysis of in vitro cultured erythroid precursors). Results The results were obtained in eight patients with the β+/β+ and β+/β0 genotypes, who were treated with a starting dosage of 1 mg/day sirolimus for 24-48 weeks. The first finding of this study was that the expression of γ-globin mRNA increased in the blood and erythroid precursor cells isolated from β-thalassemia patients treated with low-dose sirolimus. This trial also led to the important finding that sirolimus influences erythropoiesis and reduces biochemical markers associated with ineffective erythropoiesis (excess free α-globin chains, bilirubin, soluble transferrin receptor, and ferritin). A decrease in the transfusion demand index was observed in most (7/8) of the patients. The drug was well tolerated, with minor effects on the immunophenotype, and an only side effect of frequently occurring stomatitis. Conclusion The data obtained indicate that low doses of sirolimus modify hematopoiesis and induce increased expression of γ-globin genes in a subset of patients with β-thalassemia. Further clinical trials are warranted, possibly including testing of the drug in patients with less severe forms of the disease and exploring combination therapies.
Collapse
Affiliation(s)
- Cristina Zuccato
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Lucia Carmela Cosenza
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Matteo Zurlo
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Jessica Gasparello
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Chiara Papi
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Elisabetta D’Aversa
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Giulia Breveglieri
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
| | - Ilaria Lampronti
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
- Thal-LAB, Laboratorio di Ricerca Elio Zago sulla Terapia Farmacologica e Farmacogenomica della Talassemia, Università degli Studi di Ferrara, Ferrara, Italy
| | - Alessia Finotti
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
- Thal-LAB, Laboratorio di Ricerca Elio Zago sulla Terapia Farmacologica e Farmacogenomica della Talassemia, Università degli Studi di Ferrara, Ferrara, Italy
| | - Monica Borgatti
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, Ferrara, Italy
- Thal-LAB, Laboratorio di Ricerca Elio Zago sulla Terapia Farmacologica e Farmacogenomica della Talassemia, Università degli Studi di Ferrara, Ferrara, Italy
| | - Chiara Scapoli
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biologia ed Evoluzione, Università degli Studi di Ferrara, Ferrara, Italy
| | - Alice Stievano
- Unità Operativa Interdipartimentale di Day Hospital della Talassemia e delle Emoglobinopatie, Arcispedale S. Anna di Ferrara, Ferrara, Italy
| | - Monica Fortini
- Unità Operativa Interdipartimentale di Day Hospital della Talassemia e delle Emoglobinopatie, Arcispedale S. Anna di Ferrara, Ferrara, Italy
| | - Eric Ramazzotti
- Laboratorio Unico Metropolitano, Ospedale Maggiore, Azienda USL di Bologna, Bologna, Italy
| | - Nicola Marchetti
- Dipartimento di Scienze Chimiche, Farmaceutiche e Agrarie, Università degli Studi di Ferrara, Ferrara, Italy
| | | | - Maria Rita Gamberini
- Unità Operativa Interdipartimentale di Day Hospital della Talassemia e delle Emoglobinopatie, Arcispedale S. Anna di Ferrara, via Aldo Moro, 8, Ferrara 44124, Italy
| | - Roberto Gambari
- Dipartimento di Scienze della Vita e Biotecnologie, Sezione di Biochimica e Biologia Molecolare, Università degli Studi di Ferrara, via Fossato di Mortara, 74, Ferrara 44121, Italy
- Thal-LAB, Laboratorio di Ricerca Elio Zago sulla Terapia Farmacologica e Farmacogenomica della Talassemia, Università degli Studi di Ferrara, Ferrara, Italy
- Center ‘Chiara Gemmo and Elio Zago’ for the Research on Thalassemia, Università degli Studi di Ferrara, Ferrara, Italy
| |
Collapse
|
8
|
Single Nucleotide Polymorphisms in XMN1-HBG2, HBS1L-MYB, and BCL11A and Their Relation to High Fetal Hemoglobin Levels That Alleviate Anemia. Diagnostics (Basel) 2022; 12:diagnostics12061374. [PMID: 35741184 PMCID: PMC9221560 DOI: 10.3390/diagnostics12061374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
Anemia is a condition in which red blood cells and/or hemoglobin (Hb) concentrations are decreased below the normal range, resulting in a lack of oxygen being transported to tissues and organs. Those afflicted with this condition may feel lethargic and weak, which reduces their quality of life. The condition may be manifested in inherited blood disorders, such as thalassemia and sickle cell disease, whereas acquired disorders include aplastic anemia, chronic disease, drug toxicity, pregnancy, and nutritional deficiency. The augmentation of fetal hemoglobin (HbF) results in the reduction in clinical symptoms in beta-hemoglobinopathies. Several transcription factors as well as medications such as hydroxyurea may help red blood cells produce more HbF. HbF expression increases with the downregulation of three main quantitative trait loci, namely, the XMN1-HBG2, HBS1L-MYB, and BCL11A genes. These genes contain single nucleotide polymorphisms (SNPs) that modulate the expression of HbF differently in various populations. Allele discrimination is important in SNP genotyping and is widely applied in many assays. In conclusion, the expression of HbF with a genetic modifier is crucial in determining the severity of anemic diseases, and genetic modification of HbF expression may offer clinical benefits in diagnosis and disease management.
Collapse
|
9
|
Studying the some biochemical parameters for thalassemia patients in AL-Najaf province. Int J Health Sci (Qassim) 2022. [DOI: 10.53730/ijhs.v6ns4.6313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The current study aims to study some biochemical indicators for thalassemia patients in the Najaf governorate, and the study included 25 patients with major beta thalassemia during the period from November 2019 to February 2020 and their ages ranged between (2 - 65) years and 15 healthy people who arenot He had genetic blood diseases and their ages ranged between (2-65) years. The results of the study showed that there are in some biochemical indicators, as there was a significant increase in the level of the enzyme of liver function ALT that was the focus in patients 23.74 ± 29.53 U\L and in healthy people it was 4.61 ± 11.67U\L Also, the creatinine concentration patients 9.92 ± 26.08 mmol\L and in healthy was, 68.06 ± 51.54 mmol\L, Except for urea, we notice that there was a decrease in its concentration in patients 0.89 ± 3.96 mmol\L and in healthy was 1.03 ± 2.36 mmol\L, There was an increase in the concentration of iron in the blood in patients 382.05 ±64.37 Umol\L And in healthy was 9.48 ± 61.36 Umol\L, as well as a decrease in the concentration of glucose in patients and an increase in healthy people
Collapse
|
10
|
Ansari SH, Hussain Z, Zohaib M, Parveen S, Kaleem B, Qamar H, Adil O, Khan MT, Shamsi TS. A Pragmatic Scoring Tool to Predict Hydroxyurea Response Among β-Thalassemia Major Patients in Pakistan. J Pediatr Hematol Oncol 2022; 44:e77-e83. [PMID: 33710118 DOI: 10.1097/mph.0000000000002136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/02/2021] [Indexed: 11/26/2022]
Abstract
Despite high prevalence and incidence of β-thalassemia in Pakistan, there is very limited work on the use of hydroxyurea (HU) in thalassemia patients in the country. This is the first insight regarding genetic profiling of BCL11A and HU responses in Pakistani β-thalassemia. It correlates single-nucleotide polymorphisms on BCL11A (rs4671393, rs766432) and HBG2 (XmnI), age at first transfusion, and β-globin mutations with HU response in β-thalassemia major (BTM). Of 272 patients treated with HU, 98 were complete responders, 55 partial responders, and 119 nonresponders. Our analysis shows that HU response was significantly associated with patients having IVSI-1 or CD 30 mutation (P<0.001), age at first transfusion >1 year (P<0.001), and with the presence of XmnI polymorphism (P<0.001). The single-nucleotide polymorphisms of BCL11A were more prevalent among responders, but could not show significant association with HU response (P>0.05). Cumulative effect of all 5 predicting factors through simple binary scoring indicates that the likelihood of HU response increases with the number of primary and secondary genetic modifiers (P<0.001). Predictors scoring is a pragmatic tool to foresee HU response in patients with BTM. The authors recommend a score of ≥2 for starting HU therapy in Pakistani patients with BTM.
Collapse
Affiliation(s)
- Saqib H Ansari
- National Institute of Blood Diseases and Bone Marrow Transplantation
| | - Zeeshan Hussain
- National Institute of Blood Diseases and Bone Marrow Transplantation
- Omair Sana Foundation
| | - Muhammad Zohaib
- Omair Sana Foundation
- National Center for Proteomics, University of Karachi, Karachi, Pakistan
| | - Sadia Parveen
- National Institute of Blood Diseases and Bone Marrow Transplantation
| | - Bushra Kaleem
- National Institute of Blood Diseases and Bone Marrow Transplantation
| | - Hina Qamar
- National Institute of Blood Diseases and Bone Marrow Transplantation
| | | | | | - Tahir S Shamsi
- National Institute of Blood Diseases and Bone Marrow Transplantation
| |
Collapse
|
11
|
Zuccato C, Cosenza LC, Zurlo M, Lampronti I, Borgatti M, Scapoli C, Gambari R, Finotti A. Treatment of Erythroid Precursor Cells from β-Thalassemia Patients with Cinchona Alkaloids: Induction of Fetal Hemoglobin Production. Int J Mol Sci 2021; 22:13433. [PMID: 34948226 PMCID: PMC8706579 DOI: 10.3390/ijms222413433] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022] Open
Abstract
β-thalassemias are among the most common inherited hemoglobinopathies worldwide and are the result of autosomal mutations in the gene encoding β-globin, causing an absence or low-level production of adult hemoglobin (HbA). Induction of fetal hemoglobin (HbF) is considered to be of key importance for the development of therapeutic protocols for β-thalassemia and novel HbF inducers need to be proposed for pre-clinical development. The main purpose on this study was to analyze Cinchona alkaloids (cinchonidine, quinidine and cinchonine) as natural HbF-inducing agents in human erythroid cells. The analytical methods employed were Reverse Transcription quantitative real-time PCR (RT-qPCR) (for quantification of γ-globin mRNA) and High Performance Liquid Chromatography (HPLC) (for analysis of the hemoglobin pattern). After an initial analysis using the K562 cell line as an experimental model system, showing induction of hemoglobin and γ-globin mRNA, we verified whether the two more active compounds, cinchonidine and quinidine, were able to induce HbF in erythroid progenitor cells isolated from β-thalassemia patients. The data obtained demonstrate that cinchonidine and quinidine are potent inducers of γ-globin mRNA and HbF in erythroid progenitor cells isolated from nine β-thalassemia patients. In addition, both compounds were found to synergize with the HbF inducer sirolimus for maximal production of HbF. The data obtained strongly indicate that these compounds deserve consideration in the development of pre-clinical approaches for therapeutic protocols of β-thalassemia.
Collapse
Affiliation(s)
- Cristina Zuccato
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
| | - Lucia Carmela Cosenza
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
| | - Matteo Zurlo
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
| | - Ilaria Lampronti
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
- Research Laboratory “Elio Zago” on the Pharmacologic and Pharmacogenomic Therapy of Thalassemia (Thal-LAB), University of Ferrara, 44121 Ferrara, Italy
| | - Monica Borgatti
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
- Research Laboratory “Elio Zago” on the Pharmacologic and Pharmacogenomic Therapy of Thalassemia (Thal-LAB), University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Scapoli
- Section of Biology and Evolution, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy;
| | - Roberto Gambari
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
- Research Laboratory “Elio Zago” on the Pharmacologic and Pharmacogenomic Therapy of Thalassemia (Thal-LAB), University of Ferrara, 44121 Ferrara, Italy
- Interuniversity Consortium for Biotechnology (C.I.B.), 34148 Trieste, Italy
| | - Alessia Finotti
- Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy; (C.Z.); (L.C.C.); (M.Z.); (I.L.); (M.B.)
- Research Laboratory “Elio Zago” on the Pharmacologic and Pharmacogenomic Therapy of Thalassemia (Thal-LAB), University of Ferrara, 44121 Ferrara, Italy
- Interuniversity Consortium for Biotechnology (C.I.B.), 34148 Trieste, Italy
| |
Collapse
|
12
|
Musallam KM, Vitrano A, Meloni A, Addario Pollina S, Di Marco V, Hussain Ansari S, Filosa A, Ricchi P, Ceci A, Daar S, Vlachaki E, Singer ST, Naserullah ZA, Pepe A, Scondotto S, Dardanoni G, Karimi M, El-Beshlawy A, Hajipour M, Bonifazi F, Vichinsky E, Taher AT, Sankaran VG, Maggio A. Primary HBB gene mutation severity and long-term outcomes in a global cohort of β-thalassaemia. Br J Haematol 2021; 196:414-423. [PMID: 34697800 DOI: 10.1111/bjh.17897] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 01/19/2023]
Abstract
In β-thalassaemia, the severity of inherited β-globin gene mutations determines the severity of the clinical phenotype at presentation and subsequent transfusion requirements. However, data on associated long-term outcomes remain limited. We analysed data from 2109 β-thalassaemia patients with available genotypes in a global database. Genotype severity was grouped as β0 /β0 , β0 /β+ , β+ /β+ , β0 /β++ , β+ /β++ , and β++ /β++ . Patients were followed from birth until death or loss to follow-up. The median follow-up time was 34·1 years. Mortality and multiple morbidity outcomes were analyzed through five different stratification models of genotype severity groups. Interestingly, β0 and β+ mutations showed similar risk profiles. Upon adjustment for demographics and receipt of conventional therapy, patients with β0 /β0 , β0 /β+ , or β+ /β+ had a 2·104-increased risk of death [95% confidence interval (CI): 1·176-3·763, P = 0·011] and 2·956-increased odds of multiple morbidity (95% CI: 2·310-3·784, P < 0·001) compared to patients in lower genotype severity groups. Cumulative survival estimates by age 65 years were 36·8% for this subgroup compared with 90·2% for patients in lower genotype severity groups (P < 0·001). Our study identified mortality and morbidity risk estimates across various genotype severity groups in patients with β-thalassaemia and suggests inclusion of both β+ and β0 mutations in strata of greatest severity.
Collapse
Affiliation(s)
- Khaled M Musallam
- Thalassemia Center, Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Angela Vitrano
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Antonella Meloni
- MRI Unit, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | | | - Vito Di Marco
- Department of Promozione della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Saqib Hussain Ansari
- Department of Pediatric Haematology & Molecular Medicine, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Aldo Filosa
- Rare Blood Cell Disease Unit, "Cardarelli" Hospital, Naples, Italy
| | - Paolo Ricchi
- Rare Blood Cell Disease Unit, "Cardarelli" Hospital, Naples, Italy
| | - Adriana Ceci
- Fondazione per la Ricerca Farmacologica Gianni Benzi Onlus, Valenzano (BA), Italy
| | - Shahina Daar
- Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman.,Wallenberg Research Centre, Stellenbosch Institute for Advanced Study, Stellenbosch University, Stellenbosch, South Africa
| | - Efthymia Vlachaki
- Thalassaemia Unit, Ippokratio University Hospital, Thessaloniki, Greece
| | - Sylvia T Singer
- Division of Hematology-Oncology, Department of Pediatrics, University of California San Francisco, UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA
| | | | - Alessia Pepe
- MRI Unit, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | | | | | - Mehran Karimi
- Haematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amal El-Beshlawy
- Department of Pediatric Haematology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mahmoud Hajipour
- Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fedele Bonifazi
- Fondazione per la Ricerca Farmacologica Gianni Benzi Onlus, Valenzano (BA), Italy
| | - Elliott Vichinsky
- Division of Hematology-Oncology, Department of Pediatrics, University of California San Francisco, UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Aurelio Maggio
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | | |
Collapse
|
13
|
Hariharan P, Gorivale M, Sawant P, Mehta P, Nadkarni A. Significance of genetic modifiers of hemoglobinopathies leading towards precision medicine. Sci Rep 2021; 11:20906. [PMID: 34686692 PMCID: PMC8536722 DOI: 10.1038/s41598-021-00169-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 09/02/2021] [Indexed: 11/17/2022] Open
Abstract
Hemoglobinopathies though a monogenic disorder, show phenotypic variability. Hence, understanding the genetics underlying the heritable sub-phenotypes of hemoglobinopathies, specific to each population, would be prognostically useful and could inform personalized therapeutics. This study aimed to evaluate the role of genetic modifiers leading to higher HbF production with cumulative impact of the modifiers on disease severity. 200 patients (100 β-thalassemia homozygotes, 100 Sickle Cell Anemia), and 50 healthy controls were recruited. Primary screening followed with molecular analysis for confirming the β-hemoglobinopathy was performed. Co-existing α-thalassemia and the polymorphisms located in 3 genetic loci linked to HbF regulation were screened. The most remarkable result was the association of SNPs with clinically relevant phenotypic groups. The γ-globin gene promoter polymorphisms [- 158 C → T, + 25 G → A],BCL11A rs1427407 G → T, - 3 bp HBS1L-MYB rs66650371 and rs9399137 T → C polymorphisms were correlated with higher HbF, in group that has lower disease severity score (P < 0.00001), milder clinical presentation, and a significant delay in the age of the first transfusion. Our study emphasizes the complex genetic interactions underlying the disease phenotype that may be a prognostic marker for predicting the clinical severity and assist in disease management.
Collapse
Affiliation(s)
- Priya Hariharan
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th Floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, India
| | - Manju Gorivale
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th Floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, India
| | - Pratibha Sawant
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th Floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, India
| | - Pallavi Mehta
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th Floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, India
| | - Anita Nadkarni
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th Floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, India.
| |
Collapse
|
14
|
Correction of β-thalassemia by CRISPR/Cas9 editing of the α-globin locus in human hematopoietic stem cells. Blood Adv 2021; 5:1137-1153. [PMID: 33635334 DOI: 10.1182/bloodadvances.2020001996] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
β-thalassemias (β-thal) are a group of blood disorders caused by mutations in the β-globin gene (HBB) cluster. β-globin associates with α-globin to form adult hemoglobin (HbA, α2β2), the main oxygen-carrier in erythrocytes. When β-globin chains are absent or limiting, free α-globins precipitate and damage cell membranes, causing hemolysis and ineffective erythropoiesis. Clinical data show that severity of β-thal correlates with the number of inherited α-globin genes (HBA1 and HBA2), with α-globin gene deletions having a beneficial effect for patients. Here, we describe a novel strategy to treat β-thal based on genome editing of the α-globin locus in human hematopoietic stem/progenitor cells (HSPCs). Using CRISPR/Cas9, we combined 2 therapeutic approaches: (1) α-globin downregulation, by deleting the HBA2 gene to recreate an α-thalassemia trait, and (2) β-globin expression, by targeted integration of a β-globin transgene downstream the HBA2 promoter. First, we optimized the CRISPR/Cas9 strategy and corrected the pathological phenotype in a cellular model of β-thalassemia (human erythroid progenitor cell [HUDEP-2] β0). Then, we edited healthy donor HSPCs and demonstrated that they maintained long-term repopulation capacity and multipotency in xenotransplanted mice. To assess the clinical potential of this approach, we next edited β-thal HSPCs and achieved correction of α/β globin imbalance in HSPC-derived erythroblasts. As a safer option for clinical translation, we performed editing in HSPCs using Cas9 nickase showing precise editing with no InDels. Overall, we described an innovative CRISPR/Cas9 approach to improve α/β globin imbalance in thalassemic HSPCs, paving the way for novel therapeutic strategies for β-thal.
Collapse
|
15
|
Predictive SNPs for β 0-thalassemia/HbE disease severity. Sci Rep 2021; 11:10352. [PMID: 33990643 PMCID: PMC8121782 DOI: 10.1038/s41598-021-89641-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
β-Thalassemia/HbE disease has a wide spectrum of clinical phenotypes ranging from asymptomatic to dependent on regular blood transfusions. Ability to predict disease severity is helpful for clinical management and treatment decision making. A thalassemia severity score has been developed from Mediterranean β-thalassemia patients. However, different ethnic groups may have different allele frequency and linkage disequilibrium structures. Here, Thai β0-thalassemia/HbE disease genome-wild association studies (GWAS) data of 487 patients were analyzed by SNP interaction prioritization algorithm, interacting Loci (iLoci), to find predictive SNPs for disease severity. Three SNPs from two SNP interaction pairs associated with disease severity were identifies. The three-SNP disease severity risk score composed of rs766432 in BCL11A, rs9399137 in HBS1L-MYB and rs72872548 in HBE1 showed more than 85% specificity and 75% accuracy. The three-SNP predictive score was then validated in two independent cohorts of Thai and Malaysian β0-thalassemia/HbE patients with comparable specificity and accuracy. The SNP risk score could be used for prediction of clinical severity for Southeast Asia β0-thalassemia/HbE population.
Collapse
|
16
|
Wulandari RD, Lyrawati D, Fatchiyah F, Fitri LE. Thalassemia Major and Intermedia Patients in East Java do not Show Fetal Hemoglobin Level Difference in Relation to XMNI Polymorphism. Med Arch 2021; 74:90-94. [PMID: 32577047 PMCID: PMC7296421 DOI: 10.5455/medarh.2020.74.90-94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Introduction Thalassemia is a genetic disorder, which shows, varies phenotype due to genetic modifier. XmnI is one of the genetic modifiers which affect clinical severity in thalassemia. XmnI polymorphism may increase HbF production beyond fetal life, thus ameliorating the clinical phenotype. Aim this study aimed to investigate the difference in HbF level and the relation of HbF level and XmnI polymorphism in Thalassemia Major (TM) and Thalassemia Intermedia (TI) patients. Methods forty-eight beta thalassemia patients (28 males and 20 females), consists of 16 TM and 32 TI; mean age, 25.30 year old. Hemoglobin Fetal and HbA2 level were determined using High performance Liquid Chromatography (HPLC), and XmnI polymorphism was confirmed by PCR-RFLP. Statistical analysis was done using T-test, Mann-Whitney and Pearson Chi-square. Results The frequency of heterozygote (+/-) XmnI polymorphism in TM and TI patients was 56.25% vs 71.87%, while the frequency of homozygote (-/-) in TM and TI was 43.75% vs 28.13% with p value >0.05. The insignificant difference also found in HbF level between XmnI +/- and -/- in TM and TI patients. Conclusion This study revealed that thalassemia major and thalassemia intermedia patients in East Java showed similar XmnI polymorphism. These phenomena also showed by HbF level in relation to XmnI polymorphism in the phenotype groups (TM and TI).
Collapse
Affiliation(s)
- Retno Dwi Wulandari
- Doctoral Program in Medical Science, Faculty of Medicine, University of Brawijaya, Indonesia.,Department of Biomedical, Faculty of Medicine, University of Wijaya Kusuma Surabaya, Indonesia
| | - Diana Lyrawati
- Department of Pharmacy, Faculty of Medicine, University of Brawijaya, Indonesia
| | - Fatchiyah Fatchiyah
- Department of Biology, Faculty of Mathematics and Natural Sciences, University of Brawijaya, Indonesia
| | - Loeki Enggar Fitri
- Department of Parasitology, Faculty of Medicine, University of Brawijaya, Indonesia
| |
Collapse
|
17
|
Zittersteijn HA, Harteveld CL, Klaver-Flores S, Lankester AC, Hoeben RC, Staal FJT, Gonçalves MAFV. A Small Key for a Heavy Door: Genetic Therapies for the Treatment of Hemoglobinopathies. Front Genome Ed 2021; 2:617780. [PMID: 34713239 PMCID: PMC8525365 DOI: 10.3389/fgeed.2020.617780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
Throughout the past decades, the search for a treatment for severe hemoglobinopathies has gained increased interest within the scientific community. The discovery that ɤ-globin expression from intact HBG alleles complements defective HBB alleles underlying β-thalassemia and sickle cell disease, has provided a promising opening for research directed at relieving ɤ-globin repression mechanisms and, thereby, improve clinical outcomes for patients. Various gene editing strategies aim to reverse the fetal-to-adult hemoglobin switch to up-regulate ɤ-globin expression through disabling either HBG repressor genes or repressor binding sites in the HBG promoter regions. In addition to these HBB mutation-independent strategies involving fetal hemoglobin (HbF) synthesis de-repression, the expanding genome editing toolkit is providing increased accuracy to HBB mutation-specific strategies encompassing adult hemoglobin (HbA) restoration for a personalized treatment of hemoglobinopathies. Moreover, besides genome editing, more conventional gene addition strategies continue under investigation to restore HbA expression. Together, this research makes hemoglobinopathies a fertile ground for testing various innovative genetic therapies with high translational potential. Indeed, the progressive understanding of the molecular clockwork underlying the hemoglobin switch together with the ongoing optimization of genome editing tools heightens the prospect for the development of effective and safe treatments for hemoglobinopathies. In this context, clinical genetics plays an equally crucial role by shedding light on the complexity of the disease and the role of ameliorating genetic modifiers. Here, we cover the most recent insights on the molecular mechanisms underlying hemoglobin biology and hemoglobinopathies while providing an overview of state-of-the-art gene editing platforms. Additionally, current genetic therapies under development, are equally discussed.
Collapse
Affiliation(s)
- Hidde A. Zittersteijn
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis L. Harteveld
- Department of Human and Clinical Genetics, The Hemoglobinopathies Laboratory, Leiden University Medical Center, Leiden, Netherlands
| | | | - Arjan C. Lankester
- Department of Pediatrics, Stem Cell Transplantation Program, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Rob C. Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
18
|
Soremekun OS, Ezenwa C, Isewon I, Soliman M, Idowu O, Nashiru O, Fatumo S. Computational and drug target analysis of functional single nucleotide polymorphisms associated with Haemoglobin Subunit Beta (HBB) gene. Comput Biol Med 2020; 125:104018. [PMID: 33022520 DOI: 10.1016/j.compbiomed.2020.104018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/13/2020] [Accepted: 09/20/2020] [Indexed: 10/23/2022]
Abstract
There is overwhelming evidence implicating Haemoglobin Subunit Beta (HBB) protein in the onset of beta thalassaemia. In this study for the first time, we used a combined SNP informatics and computer algorithms such as Neural network, Bayesian network, and Support Vector Machine to identify deleterious non-synonymous Single Nucleotide Polymorphisms (nsSNPs) present in the HBB gene. Our findings highlight three major mutation points (R31G, W38S, and Q128P) within the HBB gene sequence that have significant statistical and computational associations with the onset of beta thalassaemia. The dynamic simulation study revealed that R31G, W38S, and Q128P elicited high structural perturbation and instability, however, the wild type protein was considerably stable. Ten compounds with therapeutic potential against HBB were also predicted by structure-based virtual screening. Interestingly, the instability caused by the mutations was reversed upon binding to a ligand. This study has been able to predict potential deleterious mutants that can be further explored in the understanding of the pathological basis of beta thalassaemia and the design of tailored inhibitors.
Collapse
Affiliation(s)
- Opeyemi S Soremekun
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Chisom Ezenwa
- Centre for Genomics Research and Innovation, National Biotechnology Agency, Nigeria
| | - Itunuoluwa Isewon
- Department of Computer and Information Sciences, Covenant University, Ota, Nigeria
| | - Mahmoud Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Omotuyi Idowu
- Centre for Genomics Research and Innovation, National Biotechnology Agency, Nigeria; Chemo-genomics Research Unit, Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Oyekanmi Nashiru
- Centre for Genomics Research and Innovation, National Biotechnology Agency, Nigeria.
| | - Segun Fatumo
- Centre for Genomics Research and Innovation, National Biotechnology Agency, Nigeria; Uganda Medical Informatics Centre and MRC/UVRI LSHTM, Uganda; London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
19
|
Kattamis A, Forni GL, Aydinok Y, Viprakasit V. Changing patterns in the epidemiology of β-thalassemia. Eur J Haematol 2020; 105:692-703. [PMID: 32886826 PMCID: PMC7692954 DOI: 10.1111/ejh.13512] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/19/2023]
Abstract
β‐thalassemia major is an inherited hemoglobinopathy that requires lifelong red blood cell transfusions and iron chelation therapy to prevent complications due to iron overload. Traditionally, β‐thalassemia has been more common in certain regions of the world such as the Mediterranean, Middle East, and Southeast Asia. However, the prevalence of β‐thalassemia is increasing in other regions, including Northern Europe and North America, primarily due to migration. This review summarizes the available data on the changing incidence and prevalence of β‐thalassemia as well as factors influencing disease frequency. The data suggest that the epidemiology of β‐thalassemia is changing: Migration has increased the prevalence of the disease in regions traditionally believed to have a low prevalence, while, at the same time, prevention and screening programs in endemic regions have reduced the number of affected individuals. Various approaches to prevention and screening have been used. Region‐specific prevention and treatment programs, customized to align with local healthcare resources and cultural values, have been effective in identifying patients and carriers and providing information and care. Significant challenges remain in universally implementing these programs.
Collapse
Affiliation(s)
- Antonis Kattamis
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Gian Luca Forni
- Centro della Microcitemia e Anemie Congenite e del Dismetabolismo del Ferro, Ospedale Galliera, Genoa, Italy
| | - Yesim Aydinok
- Department of Pediatric Hematology and Oncology, Ege University Hospital, Izmir, Turkey
| | - Vip Viprakasit
- Department of Pediatrics & Thalassemia Center, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
20
|
Ding S, Li S, Zhang S, Li Y. Genetic Alterations and Checkpoint Expression: Mechanisms and Models for Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:227-250. [PMID: 32185713 DOI: 10.1007/978-981-15-3266-5_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this chapter, we will sketch a story that begins with the breakdown of chromosome homeostasis and genomic stability. Genomic alterations may render tumor cells eternal life at the expense of immunogenicity. Although antitumor immunity can be primed through neoantigens or inflammatory signals, tumor cells have evolved countermeasures to evade immune surveillance and strike back by modulating immune checkpoint related pathways. At present, monoclonal antibody drugs targeting checkpoints like PD-1 and CTLA-4 have significantly prolonged the survival of a variety of cancer patients, and thus have marked a great achievement in the history of antitumor therapy. Nevertheless, this is not the end of the story. As the relationship between genomic alteration and checkpoint expression is being delineated though the advances of preclinical animal models and emerging technologies, novel checkpoint targets are on the way to be discovered.
Collapse
Affiliation(s)
- Shuai Ding
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Siqi Li
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Shujie Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Yan Li
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China.
| |
Collapse
|
21
|
Lal A, Bansal D. Thalassemia: Common Clinical Queries in Management. Indian J Pediatr 2020; 87:75-81. [PMID: 31620986 DOI: 10.1007/s12098-019-03065-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/20/2019] [Indexed: 01/19/2023]
Abstract
Beta thalassemia major (TM) is the most frequent form of transfusion-dependent inherited anemia in India. The thalassemia syndromes exhibit enormous variability in their genetic basis and phenotypic expression. The authors recommend that the diagnosis of TM or non-transfusion-dependent thalassemia (NTDT) should not be based on a one-time assessment. Many patients have a chronic anemia that is not severe enough to justify regular transfusions, but the clinical course can evolve with age. Continued observation may reveal that some patients who are considered NTDT will benefit from transfusions later in life. Clinical decision making can be influenced by the perceived difficulty in access to a safe blood supply and the cost of therapy. Here, authors present selected case scenarios that address common issues in the management of TM or NTDT. The recommendations are based on published evidence where available or on the authors' shared experience. Among the topics under discussion are deciding when to start regular transfusions, the role of hydroxyurea in TM, the procedure for blood administration, the use of deferasirox for chelation and monitoring of side effects, the role of splenectomy, and the prospects for gene therapy. In order to achieve an optimal outcome with blood transfusions and chelation therapy over the lifetime, it is essential to adhere to the current guidelines for the management of thalassemia.
Collapse
Affiliation(s)
- Ashutosh Lal
- Hematology/Oncology, Department of Pediatrics, University of California, San Francisco, California, USA.
| | - Deepak Bansal
- Pediatric Hematology/Oncology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
22
|
Du L, Qin D, Wang J, Yu L, Yao C, Liu L, Zhang Y, Hu T, Yuan T, Liang J, Yin A. Genetic and phenotypic analysis of a rare asymptomatic case of a homozygous Chinese Gγ +( Aγδβ) 0-thalassemia deletion in a Chinese family. Clin Biochem 2019; 76:11-16. [PMID: 31765637 DOI: 10.1016/j.clinbiochem.2019.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The clinical and hematologic features of thalassemia are due to different factors, and patients with identical genotypes may regularly exhibit variable severity. In the present work, one homozygous Chinese Gγ+(Aγδβ)0-thalassemia case with an asymptomatic phenotype, which is contrary to traditional views, was identified. Analysis of the underlying causes of this rare clinical phenotype involved accurate genetic diagnosis and detection of several genetic modifications. METHODS Six members of the proband's family were enrolled in the study. Hematological parameters and hemoglobin analysis results were recorded. A suspension-array system, multiplex gap-polymerase chain reaction (gap-PCR) and multiplex ligation-dependent probe amplification (MLPA) were used together to characterize genotypes. Sanger sequencing was utilized to examine the KLF1 gene and four primary fetal hemoglobin (Hb F)-associated single-nucleotide polymorphisms (SNPs). RESULTS Four family members carried the Chinese Gγ+(Aγδβ)0-thalassemia mutation, and a homozygous state was ultimately diagnosed for the proband. All of the Chinese Gγ+(Aγδβ)0 mutation-positive cases were coinherited with the Southern Asian α-thalassemia deletion (- - SEA/αα). Two SNP variants, rs7776054 and rs9399137, in the HBS1L-MYB locus were detected in the proband. CONCLUSIONS Thus far, this is the first study to describe the molecular characterization of a homozygous Chinese Gγ+(Aγδβ)0-thalassemia patient who exhibits no clinical symptoms. Our findings suggest that coinheritance of α-thalassemia or HBS1L-MYB locus variants may affect the clinical severity of Chinese Gγ+(Aγδβ)0-thalassemia. We conclude that the molecular examination of genetic determinants known to be associated with clinical outcomes in Chinese Gγ+(Aγδβ)0-thalassemia should be emphasized.
Collapse
Affiliation(s)
- Li Du
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Danqing Qin
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Jicheng Wang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Lihua Yu
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Cuize Yao
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Ling Liu
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Yanxia Zhang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Tingting Hu
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Tenglong Yuan
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Jie Liang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China
| | - Aihua Yin
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China; Thalassemia Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510010, China.
| |
Collapse
|
23
|
Brancaleoni V, Moukhadder HM, Consonni D, Koussa S, Di Pierro E, Cappellini MD, Taher A. Common fetal hemoglobin variants in Lebanese patients bearing the codon 29 beta gene mutation associated with different thalassemia phenotypes. Ann Hematol 2018; 98:833-840. [PMID: 30506348 DOI: 10.1007/s00277-018-3567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 11/23/2018] [Indexed: 10/27/2022]
Abstract
Beta-thalassemia can present with a wide spectrum of phenotypes determined by the coinheritance of α-thalassemia, hereditary persistence of fetal hemoglobin, and polymorphic variants in the BCL11A, HMIP, and HBB clusters. The codon 29 (cd29) mutation in the beta gene has been associated with a broad diversity of thalassemia phenotypes, possibly through genetic modifiers determining the genotype-phenotype relationship. In this study, we evaluated the effect of 10 single nucleotide polymorphisms (SNPs) on β-thalassemia severity in a group of 21 Lebanese patients bearing the cd29 mutation. Hematological parameters and clinical characteristics were evaluated according to transfusion dependence. The proportions and absolute concentrations of HbF were found to be higher in non-transfusion-dependent (NTD) patients than in transfusion-dependent (TD) ones. Iron parameters were found to be higher in TD patients. The SNPs that were evaluated included the XmnI-158 polymorphism in the HBG gene and SNPs in the BCL11A and HMIP loci. It was noted that individuals homozygous or heterozygous for the effect allele in the BCL11A and HMIP SNPs had higher HbF levels, lower ferritin concentrations, and lower liver iron content and were less likely to be transfusion dependent. Our results showed that HbF production variants may have an important impact on the severity of β-thalassemia, which might provide a severity prediction tool that can help in the anticipation of patients' phenotypes and therefore in future therapeutic decision making.
Collapse
Affiliation(s)
- Valentina Brancaleoni
- U.O.C. Medicina Generale, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Hassan M Moukhadder
- Department of Internal Medicine, American University of Beirut Medical Center, PO Box 11-0236, Beirut, 11072020, Lebanon
| | - Dario Consonni
- U.O. Epidemiologia, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Elena Di Pierro
- U.O.C. Medicina Generale, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Domenica Cappellini
- U.O.C. Medicina Generale, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Dipartimento di Scienze cliniche e di comunità, Università degli Studi di Milano, Milan, Italy
| | - Ali Taher
- Department of Internal Medicine, American University of Beirut Medical Center, PO Box 11-0236, Beirut, 11072020, Lebanon.
| |
Collapse
|
24
|
Genetic determinants related to pharmacological induction of foetal haemoglobin in transfusion-dependent HbE-β thalassaemia. Ann Hematol 2018; 98:289-299. [PMID: 30413899 DOI: 10.1007/s00277-018-3536-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 10/23/2018] [Indexed: 10/27/2022]
Abstract
Thalassaemia are the most common inherited autosomal recessive single gene disorders characterized by chronic hereditary haemolytic anaemia due to the absence or reduced synthesis of one or more of the globin chains. Haemoglobin E-β thalassaemia is the genotype responsible for approximately one half of all severe beta-thalassaemia worldwide. This study proposes to evaluate the effect of various molecular parameters on the response of hydroxyurea. Hydroxyurea was started at an initial dose of 10 mg/kg of body weight/day on 110 transfusion-dependent HbE-β thalassaemia patients. HbF level was measured by HPLC analysis. β-Thalassaemia mutations, XmnI and five other SNPs, and α-globin gene deletions and triplications were detected by ARMS-PCR, RFLP-PCR and Gap-PCR, respectively. Based on the factors for evaluating hydroxyurea-response, 42 patients were responders as they showed an increment of Hb from a mean baseline value of 6.45 g/dl (± 0.70) to 7.78 g/dl (± 0.72) post-therapy. Based on increase in HbF above the median value (14.72%) post-therapy, 78 patients were found to be responders. All the 78 responders showed mean decrease in transfusion of 74.26% (± 8.32) with a maximum decrease of 98.43%. There was a significant correlation between decrease in transfusion and increase in HbF level for all 78 responders. XmnI polymorphism showed the strongest association (p < 0.0001) with increase in HbF levels and Hb levels. Patients with α-globin gene deletions were better responders. It was concluded that hydroxyurea treatment is effective in transfusion-dependent HbE-β thalassaemia patients and the response is best in patients having both XmnI polymorphism and α-deletion.
Collapse
|
25
|
Scheps KG, Hasenahuer MA, Parisi G, Targovnik HM, García E, Veber ES, Crisp R, Elena G, Varela V, Fornasari MS. Two novel unstable hemoglobin variants due to in-frame deletions of key amino acids in the β-globin chain. Eur J Haematol 2018; 100:529-535. [DOI: 10.1111/ejh.13029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Karen G. Scheps
- Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética; Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
- Instituto de Inmunología; Genética y Metabolismo (INIGEM); CONICET-Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
| | - Marcia Anahí Hasenahuer
- Departamento de Ciencia y Tecnología; Universidad Nacional de Quilmes; Bernal, Buenos Aires Argentina
| | - Gustavo Parisi
- Departamento de Ciencia y Tecnología; Universidad Nacional de Quilmes; Bernal, Buenos Aires Argentina
| | - Héctor M. Targovnik
- Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética; Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
- Instituto de Inmunología; Genética y Metabolismo (INIGEM); CONICET-Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
| | - Eliana García
- Sección Hematología y Oncología; Servicio de Pediatría; Hospital Nacional A. Posadas; El Palomar Buenos Aires Argentina
| | - Ernesto Samuel Veber
- Servicio de Hemato-Oncología Pediátrica; Hospital General de Niños “Dr. Pedro de Elizalde”; Ciudad Autónoma de Buenos Aires Argentina
| | - Renée Crisp
- Sección Hematología y Oncología; Servicio de Pediatría; Hospital Nacional A. Posadas; El Palomar Buenos Aires Argentina
| | - Graciela Elena
- Servicio de Hemato-Oncología Pediátrica; Hospital General de Niños “Dr. Pedro de Elizalde”; Ciudad Autónoma de Buenos Aires Argentina
| | - Viviana Varela
- Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética; Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
- Instituto de Inmunología; Genética y Metabolismo (INIGEM); CONICET-Universidad de Buenos Aires; Ciudad Autónoma de Buenos Aires Argentina
| | - María Silvina Fornasari
- Departamento de Ciencia y Tecnología; Universidad Nacional de Quilmes; Bernal, Buenos Aires Argentina
| |
Collapse
|
26
|
An Aγ-globin G->A gene polymorphism associated with β 039 thalassemia globin gene and high fetal hemoglobin production. BMC MEDICAL GENETICS 2017; 18:93. [PMID: 28851297 PMCID: PMC5575872 DOI: 10.1186/s12881-017-0450-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 08/14/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increase of the expression of γ-globin gene and high production of fetal hemoglobin (HbF) in β-thalassemia patients is widely accepted as associated with a milder or even asymptomatic disease. The search for HbF-associated polymorphisms (such as the XmnI, BCL11A and MYB polymorphisms) has recently gained great attention, in order to stratify β-thalassemia patients with respect to expectancy of the first transfusion, need for annual intake of blood, response to HbF inducers (the most studied of which is hydroxyurea). METHODS Aγ-globin gene sequencing was performed on genomic DNA isolated from a total of 75 β-thalassemia patients, including 31 β039/β039, 33 β039/β+IVSI-110, 9 β+IVSI-110/β+IVSI-110, one β0IVSI-1/β+IVSI-6 and one β039/β+IVSI-6. RESULTS The results show that the rs368698783 polymorphism is present in β-thalassemia patients in the 5'UTR sequence (+25) of the Aγ-globin gene, known to affect the LYAR (human homologue of mouse Ly-1 antibody reactive clone) binding site 5'-GGTTAT-3'. This Aγ(+25 G->A) polymorphism is associated with the Gγ-globin-XmnI polymorphism and both are linked with the β039-globin gene, but not with the β+IVSI-110-globin gene. In agreement with the expectation that this mutation alters the LYAR binding activity, we found that the Aγ(+25 G->A) and Gγ-globin-XmnI polymorphisms are associated with high HbF in erythroid precursor cells isolated from β039/β039 thalassemia patients. CONCLUSIONS As a potential explanation of our findings, we hypothesize that in β-thalassemia the Gγ-globin-XmnI/Aγ-globin-(G->A) genotype is frequently under genetic linkage with β0-thalassemia mutations, but not with the β+-thalassemia mutation here studied (i.e. β+IVSI-110) and that this genetic combination has been selected within the population of β0-thalassemia patients, due to functional association with high HbF. Here we describe the characterization of the rs368698783 (+25 G->A) polymorphism of the Aγ-globin gene associated in β039 thalassemia patients with high HbF in erythroid precursor cells.
Collapse
|
27
|
MAHDAVI MR, POURFARZAD F, KOSARYAN M, AKBARI MT. In Vitro Hb Production in B-thalassemia Patients Is Not a Predictor of Clinical Responsiveness to Hydroxyurea. IRANIAN JOURNAL OF PUBLIC HEALTH 2017; 46:948-956. [PMID: 28845406 PMCID: PMC5563877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The hematologic response to hydroxyurea (HU) is varied among β-thalassemia (BT) patients. The BCL11A and SOX6 genes are involved in response to HU. This study aimed to investigate the in-vitro responsiveness of HU among BT major patients homozygote for IVSII-1G>A mutation and XmnI single nucleotide polymorphism (SNP) in order to find whether the in-vitro Hb concentration is a predictor of clinical (HU) responsiveness. METHODS In this case-control study, twenty BT patients homozygote for IVSII-1G>A mutation and XmnI SNP from Thalassemia Research Center, Sari, Iran in 2015 were selected and categorized into two groups of 10 Responder (R) and 10 Non-Responder (NR) according to their clinical HU response. Ten healthy individuals as a control group were also selected. Hematopoietic erythroid progenitors were expanded from peripheral blood. Hb concentration was measured using photometry method. The flow cytometry and real-time PCR methods were applied for the analysis of cell surface markers (CD71 and CD235a) and gene expression (BCL11A and SOX6), respectively. RESULTS R and NR groups produced higher amount of Basic Hb than C group in cell culture medium at day 14 (P<0.05). After HU treatment, in R group, Hb levels was significantly elevated in comparison to NR and C group (P<0.05). BCL11A expression was decreased after exposure to HU in all groups while SOX6 expression was only down-regulated in C group, and its expression was increased in R and NR groups after HU treatment. CONCLUSION Since different factors including wide networks of intracellular factors and individual differences between patients can affect response to HU in patients, the increasing Hemoglobin on culture medium alone cannot predict clinical responsiveness to that drug.
Collapse
|
28
|
Molecular basis of β thalassemia and potential therapeutic targets. Blood Cells Mol Dis 2017; 70:54-65. [PMID: 28651846 DOI: 10.1016/j.bcmd.2017.06.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/09/2017] [Accepted: 06/09/2017] [Indexed: 12/15/2022]
Abstract
The remarkable phenotypic diversity of β thalassemia that range from severe anemia and transfusion-dependency, to a clinically asymptomatic state exemplifies how a spectrum of disease severity can be generated in single gene disorders. While the genetic basis for β thalassemia, and how severity of the anemia could be modified at different levels of its pathophysiology have been well documented, therapy remains largely supportive with bone marrow transplant being the only cure. Identification of the genetic variants modifying fetal hemoglobin (HbF) production in combination with α globin genotype provide some prediction of disease severity for β thalassemia but generation of a personalized genetic risk score to inform prognosis and guide management requires a larger panel of genetic modifiers yet to be discovered. Nonetheless, genetic studies have been successful in characterizing the key variants and pathways involved in HbF regulation, providing new therapeutic targets for HbF reactivation. BCL11A has been established as a quantitative repressor, and progress has been made in manipulating its expression using genomic and gene-editing approaches for therapeutic benefits. Recent discoveries and understanding in the mechanisms associated with ineffective and abnormal erythropoiesis have also provided additional therapeutic targets, a couple of which are currently being tested in clinical trials.
Collapse
|
29
|
Traeger-Synodinos J, Harteveld CL. Preconception carrier screening and prenatal diagnosis in thalassemia and hemoglobinopathies: challenges and future perspectives. Expert Rev Mol Diagn 2017; 17:281-291. [DOI: 10.1080/14737159.2017.1285701] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Joanne Traeger-Synodinos
- Department of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, Athens, Greece
| | - Cornelis L. Harteveld
- Department of Clinical Genetics, Laboratory for Diagnostic Genome Analysis (LDGA), Leiden, The Netherlands
| |
Collapse
|
30
|
Thein SL. Genetic Basis and Genetic Modifiers of β-Thalassemia and Sickle Cell Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1013:27-57. [PMID: 29127676 DOI: 10.1007/978-1-4939-7299-9_2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
β-thalassemia and sickle cell disease (SCD) are prototypical Mendelian single gene disorders, both caused by mutations affecting the adult β-globin gene. Despite the apparent genetic simplicity, both disorders display a remarkable spectrum of phenotypic severity and share two major genetic modifiers-α-globin genotype and innate ability to produce fetal hemoglobin (HbF, α2γ2).This article provides an overview of the genetic basis for SCD and β-thalassemia, and genetic modifiers identified through phenotype correlation studies. Identification of the genetic variants modifying HbF production in combination with α-globin genotype provide some prediction of disease severity for β-thalassemia and SCD but generation of a personalized genetic risk score to inform prognosis and guide management requires a larger panel of genetic modifiers yet to be discovered.Nonetheless, genetic studies have been successful in characterizing some of the key variants and pathways involved in HbF regulation, providing new therapeutic targets for HbF reactivation.
Collapse
Affiliation(s)
- Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Building 10, Room 6S241 MSC 1589, 10 Center Dr., Bethesda, MD, 20892-1589, USA.
| |
Collapse
|
31
|
Bianchi N, Cosenza LC, Lampronti I, Finotti A, Breveglieri G, Zuccato C, Fabbri E, Marzaro G, Chilin A, De Angelis G, Borgatti M, Gallucci C, Alfieri C, Ribersani M, Isgrò A, Marziali M, Gaziev J, Morrone A, Sodani P, Lucarelli G, Gambari R, Paciaroni K. Structural and Functional Insights on an Uncharacterized Aγ-Globin-Gene Polymorphism Present in Four β0-Thalassemia Families with High Fetal Hemoglobin Levels. Mol Diagn Ther 2016; 20:161-73. [PMID: 26897028 DOI: 10.1007/s40291-016-0187-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Several DNA polymorphisms have been associated with high production of fetal hemoglobin (HbF), although the molecular basis is not completely understood. In order to identify and characterize novel HbF-associated elements, we focused on five probands and their four families (from Egypt, Iraq and Iran) with thalassemia major (either β(0)-IVSII-1 or β(0)-IVSI-1) and unusual HbF elevation (>98 %), congenital or acquired after rejection of bone marrow transplantation, suggesting an anticipated favorable genetic background to high HbF expression. METHODS Patient recruitment, genomic DNA sequencing, western blotting, electrophoretic mobility shift assays, surface plasmon resonance (SPR) biospecific interaction analysis, bioinformatics analyses based on docking experiments. RESULTS A polymorphism of the Aγ-globin gene is here studied in four families with β(0)-thalassemia (β(0)-IVSII-1 and β(0)-IVSI-1) and expressing unusual high HbF levels, congenital or acquired after rejection of bone marrow transplantation. This (G→A) polymorphism is present at position +25 of the Aγ-globin genes, corresponding to a 5'-UTR region of the Aγ-globin mRNA and, when present, is physically linked in chromosomes 11 of all the familiar members studied to the XmnI polymorphism and to the β(0)-thalassemia mutations. The region corresponding to the +25(G→A) polymorphism of the Aγ-globin gene belongs to a sequence recognized by DNA-binding protein complexes, including LYAR (Ly-1 antibody reactive clone), a zinc-finger transcription factor previously proposed to be involved in down-regulation of the expression of γ-globin genes in erythroid cells. CONCLUSION We found a novel polymorphism of the Aγ-globin gene in four families with β(0)-thalassemia and high levels of HbF expression. Additionally, we report evidence suggesting that the Aγ-globin gene +25(G→A) polymorphism decreases the efficiency of the interaction between this sequence and specific DNA binding protein complexes.
Collapse
Affiliation(s)
- Nicoletta Bianchi
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | | | - Ilaria Lampronti
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Giulia Breveglieri
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Cristina Zuccato
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Enrica Fabbri
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Adriana Chilin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Gioia De Angelis
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy
| | - Cristiano Gallucci
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Cecilia Alfieri
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Michela Ribersani
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Antonella Isgrò
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Marco Marziali
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Javid Gaziev
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Aldo Morrone
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Pietro Sodani
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Guido Lucarelli
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, Ferrara, Italy.
- Biotechnology Center, Ferrara University, Ferrara, Italy.
| | - Katia Paciaroni
- International Centre for Transplantation in Thalassemia and Sickle Cell Anaemia, Mediterranean Institute of Haematology, Policlinic of "Tor Vergata" University, Rome, Italy
| |
Collapse
|
32
|
Gallagher PG, Maksimova Y, Schulz VP, Forget BG. Mutation in a Highly Conserved COOH-Terminal Residue of Krüppel-Like Factor 1 Associated with Elevated Hb F in a Compound Heterozygous β-Thalassemia Patient with a Nontransfusion-Dependent Thalassemia Phenotype. Hemoglobin 2016; 40:361-364. [PMID: 27821015 DOI: 10.1080/03630269.2016.1214921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We present a patient with a compound heterozygosity codon 39 (C > T) (β0) [or β39(C5)Gln→Stop (G39X); CAG > TAG; HBB: c.118C > T] and -87 (C > T) (β+) (HBB: c.-137C > T) β-globin mutations, a non transfusion-dependent thalassemia phenotype and 97.0% fetal hemoglobin. A novel heterozygous mutation was identified in a highly conserved residue in the COOH-terminus of the Krüppel-like factor 1, R360H, that likely altered DNA-binding and impaired transactivation.
Collapse
Affiliation(s)
- Patrick G Gallagher
- a Department of Pediatrics , Yale University School of Medicine , New Haven , CT , USA.,b Departments of Pathology and Genetics , University School of Medicine , New Haven , CT , USA
| | - Yelena Maksimova
- a Department of Pediatrics , Yale University School of Medicine , New Haven , CT , USA
| | - Vincent P Schulz
- a Department of Pediatrics , Yale University School of Medicine , New Haven , CT , USA
| | - Bernard G Forget
- c Department of Internal Medicine , Yale School of Medicine , New Haven , CT , USA
| |
Collapse
|
33
|
Finotti A, Borgatti M, Bianchi N, Zuccato C, Lampronti I, Gambari R. Orphan Drugs and Potential Novel Approaches for Therapies of β-Thalassemia: Current Status and Future Expectations. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1135793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Mettananda S, Gibbons RJ, Higgs DR. Understanding α-globin gene regulation and implications for the treatment of β-thalassemia. Ann N Y Acad Sci 2015; 1368:16-24. [PMID: 26695885 DOI: 10.1111/nyas.12988] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over the past three decades, a vast amount of new information has been uncovered describing how the globin genes are regulated. This knowledge has provided significant insights into the general understanding of the regulation of human genes. It is now known that molecular defects within and around the α- and β-globin genes, as well as in the distant regulatory elements, can cause thalassemia. Unbalanced production of globin chains owing to defective synthesis of one, and the continued unopposed synthesis of another, is the central causative factor in the cellular pathology and pathophysiology of thalassemia. A large body of clinical, genetic, and experimental evidence suggests that altering globin chain imbalance by reducing the production of α-globin synthesis ameliorates the disease severity in patients with β-thalassemia. With the development of new genetic-based therapeutic tools that have a potential to decrease the expression of a selected gene in a tissue-specific manner, the possibility of decreasing expression of the α-globin gene to improve the clinical severity of β-thalassemia could become a reality.
Collapse
Affiliation(s)
- Sachith Mettananda
- Medical Research Council (MRC) Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Pediatrics, Faculty of Medicine, University of Kelaniya, Sri Lanka
| | - Richard J Gibbons
- Medical Research Council (MRC) Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Douglas R Higgs
- Medical Research Council (MRC) Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Oxford National Institute for Health Research Biomedical Research Centre, Blood Theme, Oxford University Hospital, Oxford, United Kingdom
| |
Collapse
|
35
|
Breveglieri G, Finotti A, Borgatti M, Gambari R. Recent patents and technology transfer for molecular diagnosis of β-thalassemia and other hemoglobinopathies. Expert Opin Ther Pat 2015; 25:1453-76. [PMID: 26413795 DOI: 10.1517/13543776.2015.1090427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Biological tests and genetic analyses for diagnosis and characterization of hematological diseases in health laboratories are designed with the aim of meeting the major medical needs of hospitals and pharmaceutical companies involved in this field of applied biomedicine. Genetic testing approaches to perform diagnosis consist of molecular techniques, which should be absolutely reproducible, fast, sensitive, cheap, and portable. AREAS COVERED Biological tests analyzed involve adult/newborn subjects, whereas genetic analyses involve adult thalassemia patients, newborns, embryos/fetuses (including non-invasive prenatal diagnosis), pre-implantation embryos, and pre-fertilization oocytes. EXPERT OPINION The most recent findings in the diagnostic approach for β-thalassemias are related to three major fields of investigation: moving towards ultrasensitive methodologies for effective detection of the primary causative mutation of β-thalassemia, including the development of polymerase chain reaction-free approaches and non-invasive prenatal diagnosis; comparing analyses of the genotype of β-thalassemia patients to high-HbF-associated polymorphisms; introducing whole genome association assays and next-generation sequencing. All these issues should be considered and discussed in the context of several aspects, including regulatory, ethical and social issues. DNA sequence data aligned with the identification of genes central to the induction, development, progression, and outcome of β-thalassemia will be a key point for directing personalized therapy.
Collapse
Affiliation(s)
- Giulia Breveglieri
- a 1 University of Ferrara, Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology , Ferrara, Italy
| | - Alessia Finotti
- a 1 University of Ferrara, Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology , Ferrara, Italy
| | - Monica Borgatti
- a 1 University of Ferrara, Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology , Ferrara, Italy
| | - Roberto Gambari
- b 2 University of Ferrara, Section of Biochemistry and Molecular Biology, Department of Life Sciences and Biotechnology , Via Fossato di Mortara 74, 44121 Ferrara, Italy +39 05 32 97 44 43 ; +39 05 32 97 45 00 ;
| |
Collapse
|
36
|
Motovali-Bashi M, Ghasemi T. Role of XmnIgG Polymorphism in Hydroxyurea Treatment and Fetal Hemoglobin Level at Isfahanian Intermediate β-Thalassemia Patients. IRANIAN BIOMEDICAL JOURNAL 2015; 19:177-82. [PMID: 26024726 PMCID: PMC4571014 DOI: 10.7508/ibj.2015.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND β-thalassemia is the most common monogenic disorder in human. The (C-->T) polymorphism at -158 upstream region of the γG-globin gene and pharmacological factors such as hydroxyurea have been reported to influence γ-globin gene expression and the severity of clinical symptoms of β-thalassemia. METHODS In the present study, 51 β-thalassemia intermediate patients were studied. Xmn1γG polymorphism genotype was determined using Tetra-Primer ARMS-PCR technique. Hemoglobin (Hb) and fetal hemoglobin (HbF) levels were determined by gel electrophoresis. RESULTS Of 51 patients, 35 (68.6%) patients were heterozygous (CT) and 16 (31.4%) patients were homozygous (CC). Of 30 patients under treatment by hydroxyurea, 20 (66.7%) patients were heterozygous (CT) and 10 (33.3%) patients were homozygous (CC). Our results demonstrated that in the heterozygous (CT) genotype, the Hb (9.58 ± 1.25 gm/dl) and HbF (89.30 ± 21.87) levels were significantly higher in comparison with homozygous (CC) genotype (7.94 ± 1.34 gm/dl and 70.32 ± 40.56, respectively). Furthermore, we observed that after drug usage, the Hb and HbF levels in patients with heterozygous (CT) genotype (0.7 ± 1.26 gm/dl and 5.95 ± 14.8, respectively) raised more in comparison with homozygous (CC) genotype (0.26 ± 1.43 gm/dl and 0.8 ± 1.31, respectively). CONCLUSION Hb and HbF levels in the patients carrying T allele are increased significantly, and they also response to hydroxyurea treatment.
Collapse
Affiliation(s)
- Majid Motovali-Bashi
- Genetic Division, Dept. of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Tayyebeh Ghasemi
- Genetic Division, Dept. of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
37
|
α-Globin as a molecular target in the treatment of β-thalassemia. Blood 2015; 125:3694-701. [PMID: 25869286 DOI: 10.1182/blood-2015-03-633594] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/31/2015] [Indexed: 12/31/2022] Open
Abstract
The thalassemias, together with sickle cell anemia and its variants, are the world's most common form of inherited anemia, and in economically undeveloped countries, they still account for tens of thousands of premature deaths every year. In developed countries, treatment of thalassemia is also still far from ideal, requiring lifelong transfusion or allogeneic bone marrow transplantation. Clinical and molecular genetic studies over the course of the last 50 years have demonstrated how coinheritance of modifier genes, which alter the balance of α-like and β-like globin gene expression, may transform severe, transfusion-dependent thalassemia into relatively mild forms of anemia. Most attention has been paid to pathways that increase γ-globin expression, and hence the production of fetal hemoglobin. Here we review the evidence that reduction of α-globin expression may provide an equally plausible approach to ameliorating clinically severe forms of β-thalassemia, and in particular, the very common subgroup of patients with hemoglobin E β-thalassemia that makes up approximately half of all patients born each year with severe β-thalassemia.
Collapse
|
38
|
Muszlak M, Pissard S, Badens C, Chamouine A, Maillard O, Thuret I. Genetic Modifiers of Sickle Cell Disease: A Genotype-Phenotype Relationship Study in a Cohort of 82 Children on Mayotte Island. Hemoglobin 2015; 39:156-61. [DOI: 10.3109/03630269.2015.1023897] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Al-Allawi NAS, Puehringer H, Raheem RA, Oberkanins C. Genetic Modifiers in β-Thalassemia Intermedia: A Study on 102 Iraqi Arab Patients. Genet Test Mol Biomarkers 2015; 19:242-7. [PMID: 25751242 DOI: 10.1089/gtmb.2014.0310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To determine the molecular basis of β-thalassemia intermedia (TI) and the contribution of the three hemoglobin F (HbF) quantitative trait loci (QTLs) on chromosomes 11, 2, and 6 to the milder phenotype, a total of 102 Iraqi Arab patients with TI were studied. The β and α genotypes as well as HBG2 g. 158 C>T (rs7482144), BCL11A (rs1427407 and rs10189857), and HBS1L-MYB (rs28384513 and rs9399137) by multiplex polymerase chain reaction and reverse hybridization were studied. A total of 21 different β-thalassemia mutations arranged in 35 different genotypes were identified. The genotypes encompassed β(+)/β(+) mutations in 33 cases, β(+)/β(0) in 17 cases, β(0)/β(0) in 47 cases, β(0)/wild type in 3 and β(0)/Hb E in 2 cases. The most common was IVS-II-1 (G>A)/IVS-II-1 (G>A), followed by IVS-I-6 (T>C)/IVS-I-6 (T>C) and IVS-I-110 (G>A)/IVS-I-110 (G>A), in 31.4%, 17.6%, and 6.9%, respectively. Alpha-thalassemia mutations were found in 15.2% of those homozygous for the β-mutations, while α gene triplication was identified in all three heterozygotes. Of the five QTLs tested, only rs7482144 and rs10189857 were significantly associated with β(0)/β(0) when compared to β(+)/β(+), with odds ratios of 6.4 (95% confidence interval [CI] 2.9-14.0) and 3.2 (95% CI 1.2-8.6), respectively. In conclusion, this study has demonstrated that among Iraqi patients with thal intermedia, the main contributors to the milder phenotype were β(+) alleles, XmnI polymorphism, and BCL11A (rs10189857), while other QTLs on chromosomes 2 and 6, as well as alpha-thalassemia, were not significantly relevant.
Collapse
Affiliation(s)
- Nasir A S Al-Allawi
- 1 Faculty of Medical Sciences, Scientific Research Center, University of Duhok , Duhok, Iraq
| | | | | | | |
Collapse
|
40
|
Ali N, Ayyub M, Khan SA, Ahmed S, Abbas K, Malik HS, Tashfeen S. Frequency of Gγ-globin promoter −158 (C>T) XmnI polymorphism in patients with homozygous/compound heterozygous beta thalassaemia. Hematol Oncol Stem Cell Ther 2015; 8:10-5. [DOI: 10.1016/j.hemonc.2014.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 11/20/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022] Open
|
41
|
Danjou F, Francavilla M, Anni F, Satta S, Demartis FR, Perseu L, Manca M, Sollaino MC, Manunza L, Mereu E, Marceddu G, Pissard S, Joly P, Thuret I, Origa R, Borg J, Forni GL, Piga A, Lai ME, Badens C, Moi P, Galanello R. A genetic score for the prediction of beta-thalassemia severity. Haematologica 2014; 100:452-7. [PMID: 25480500 DOI: 10.3324/haematol.2014.113886] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Clinical and hematologic characteristics of beta(β)-thalassemia are determined by several factors resulting in a wide spectrum of severity. Phenotype modulators are: HBB mutations, HBA defects and fetal hemoglobin production modulators (HBG2:g.-158C>T polymorphism, HBS1L-MYB intergenic region and the BCL11A). We characterized 54 genetic variants at these five loci robustly associated with the amelioration of beta-thalassemia phenotype, to build a predictive score of severity using a representative cohort of 890 β-thalassemic patients. Using Cox proportional hazard analysis on a training set, we assessed the effect of these loci on the age at which patient started regular transfusions, built a Thalassemia Severity Score, and validated it on a testing set. Discriminatory power of the model was high (C-index=0.705; R(2)=0.343) and the validation conducted on the testing set confirmed its predictive accuracy with transfusion-free survival probability (P<0.001) and with transfusion dependency status (Area Under the Receiver Operating Characteristic Curve=0.774; P<0.001). Finally, an automatized on-line calculation of the score was made available at http://tss.unica.it. Besides the accurate assessment of genetic predictors effect, the present results could be helpful in the management of patients, both as a predictive score for screening and a standardized scale of severity to overcome the major-intermedia dichotomy and support clinical decisions.
Collapse
Affiliation(s)
- Fabrice Danjou
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Marcella Francavilla
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Franco Anni
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Stefania Satta
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Franca-Rosa Demartis
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Lucia Perseu
- Ospedale Regionale per le Microcitemie, ASL8, Cagliari, Italy
| | - Matteo Manca
- Ospedale Regionale per le Microcitemie, ASL8, Cagliari, Italy
| | | | - Laura Manunza
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Elisabetta Mereu
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Giuseppe Marceddu
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Serge Pissard
- Laboratoire de Génétique, Hôpital Henri Mondor, Créteil, France
| | - Philippe Joly
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpital Edouard Herriot, Lyon, France
| | - Isabelle Thuret
- French reference center for Thalassemia and Service d'Onco-Hématologie Pédiatrique, Assistance Publique Hôpitaux de Marseille, France
| | - Raffaella Origa
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences and Laboratory of Molecular Genetics, Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Gian Luca Forni
- Ematologia - Centro della Microcitemia ed Anemie Congenite, Ospedale Galliera, Genova, Italy
| | | | | | - Catherine Badens
- French reference center for Thalassemia and Department of Genetics, Assistance Publique Hopitaux de Marseille and Aix-Marseille University, France
| | - Paolo Moi
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy Ospedale Regionale per le Microcitemie, ASL8, Cagliari, Italy
| | - Renzo Galanello
- Dipartimento di Sanità Pubblica, Medicina Clinica e Molecolare, Università degli Studi di Cagliari, Italy Ospedale Regionale per le Microcitemie, ASL8, Cagliari, Italy
| |
Collapse
|
42
|
Prise en charge actuelle des thalassémies intermédiaires. Transfus Clin Biol 2014; 21:143-9. [DOI: 10.1016/j.tracli.2014.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/19/2022]
|
43
|
Yu LH, Liu D, Cai R, Shang X, Zhang XH, Ma XX, Yan SH, Fang P, Zheng CG, Wei XF, Liu YH, Zhou TB, Xu XM. Changes in hematological parameters in α-thalassemia individuals co-inherited with erythroid Krüppel-like factor mutations. Clin Genet 2014; 88:56-61. [PMID: 24930900 DOI: 10.1111/cge.12443] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 05/28/2014] [Accepted: 06/11/2014] [Indexed: 12/20/2022]
Abstract
Phenotypic variations in α-thalassemia mainly depend on the defective α-globin gene number. Genetic modifiers of the phenotype of Hemoglobin H (HbH) disease were poorly reported, apart from β-thalassemia allele that was identified ameliorating the severity of α-thalassemia. Because erythroid Krüppel-like factor (KLF1) mutations can modulate the red blood phenotype, we evaluated its effect on the α-thalassemia phenotype. Overall, we identified 72 subjects with five different KLF1 heterozygous mutations in 1468 individuals, including 65 out of 432 α-thalassemia carriers with fetal hemoglobin (HbF) levels ≥1%, 0 out of 310 carriers with HbF levels <1% and 7 out of 726 HbH disease patients. We firstly established the link between KLF1 mutations and relatively elevated hemoglobin A2 (HbA2 ) and HbF levels, along with lower mean corpuscular volume (MCV) and mean corpuscular hemoglobin (MCH) values in a group of α-thalassemia carriers. However, we concluded that KLF1 mutations were not significantly linked to HbH disease severity. On the basis of HBA or HBB genotype and gender, clinical severity of patients with HbH disease was correctly predicted in 73.3% cases. It may improve the screening and diagnostic assessment of α-thalassemia.
Collapse
Affiliation(s)
- L-H Yu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - D Liu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - R Cai
- Department of Birth Health and Heredity, Liuzhou Women and Children Care Hospital, Liuzhou, Guangxi, P.R.China
| | - X Shang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - X-H Zhang
- Department of Hematology, 303rd Hospital of the People's Liberation Army, Nanning, Guangxi, P.R.China
| | - X-X Ma
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - S-H Yan
- Laboratory of Medical Genetics, Maternal and Child Health Care Hospital of Qinzhou, Qinzhou, Guangxi, P.R.China
| | - P Fang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - C-G Zheng
- Prenatal Diagnostic Center, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning, Guangxi, P.R.China
| | - X-F Wei
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| | - Y-H Liu
- Prenatal Diagnosis Center, Maternal and Child Health Hospital, Dongguan, Guangdong, P.R.China
| | - T-B Zhou
- Clinical Laboratory, Yunnan Women and Children Care Hospital, Kunming, Yunnan, P.R.China
| | - X-M Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R.China
| |
Collapse
|
44
|
Traeger-Synodinos J, Harteveld CL. Advances in technologies for screening and diagnosis of hemoglobinopathies. Biomark Med 2014; 8:119-31. [PMID: 24325233 DOI: 10.2217/bmm.13.103] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hemoglobinopathies constitute the most common monogenic disorders worldwide, caused by mutations in the globin genes that synthesize the globin chains of hemoglobin. Synthesis may be reduced (thalassemia) or underlie abnormal hemoglobins. Mutation interactions produce a wide range of disorders. For neonatal and antenatal screening, identification of affected newborns or carriers is achieved by hematological tests. DNA analysis supports definitive diagnosis, and additionally facilitates prenatal diagnosis procedures. Most methods used today have been developed over several decades, with few recent advances in hematology methods. However, DNA methods evolve continuously. With global migration and multiethnic societies the trend is from targeted, population-specific methods towards generic methods, such as Sanger sequencing (point mutations) and multiplex ligation probe amplification (deletions). DNA microarrays constitute an advanced DNA method for some mutation categories. The newest DNA technology is next-generation sequencing. Although not completely ready for routine use currently, next-generation sequencing may soon become a reality for some hemoglobin diagnostic laboratories.
Collapse
Affiliation(s)
- Jan Traeger-Synodinos
- Medical Genetics, University of Athens, St Sophia's Children's Hospital, Athens 11527, Greece.
| | | |
Collapse
|
45
|
Abstract
Thalassemia is the most common monogenic inherited disease worldwide and it affects most countries to various extents. This review summarizes the current approaches to phenotypic and genotypic diagnosis of thalassemia in clinical practice. Prevention strategies that encompass carrier screening, genetic counseling and prenatal diagnosis are discussed. The importance of public education and an awareness of a changing perception regarding this group of diseases are emphasized. It also addresses the impact of the rapidly increasing knowledge in disease severity modification by hemoglobin F (Hb F).
Collapse
Affiliation(s)
- Ho-Wan Ip
- Department of Pathology and Clinical Biochemistry, Queen Mary Hospital , Hong Kong SAR , China and
| | | |
Collapse
|
46
|
Pakdee N, Yamsri S, Fucharoen G, Sanchaisuriya K, Pissard S, Fucharoen S. Variability of hemoglobin F expression in hemoglobin EE disease: Hematological and molecular analysis. Blood Cells Mol Dis 2014; 53:11-5. [DOI: 10.1016/j.bcmd.2014.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/01/2014] [Indexed: 12/17/2022]
|
47
|
Fanis P, Kousiappa I, Phylactides M, Kleanthous M. Genotyping of BCL11A and HBS1L-MYB SNPs associated with fetal haemoglobin levels: a SNaPshot minisequencing approach. BMC Genomics 2014; 15:108. [PMID: 24502199 PMCID: PMC3922441 DOI: 10.1186/1471-2164-15-108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/24/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND B-thalassaemia and sickle cell disease (SCD) are two of the most common monogenic diseases that are found in many populations worldwide. In both disorders the clinical severity is highly variable, with the persistence of fetal haemoglobin (HbF) being one of the major ameliorating factors. HbF levels are affected by, amongst other factors, single nucleotide polymorphisms (SNPs) at the BCL11A gene and the HBS1L-MYB intergenic region, which are located outside the β-globin locus. For this reason, we developed two multiplex assays that allow the genotyping of SNPs at these two genomic regions which have been shown to be associated with variable HbF levels in different populations. RESULTS Two multiplex assays based on the SNaPshot minisequencing approach were developed. The two assays can be used to simultaneous genotype twelve SNPs at the BCL11A gene and sixteen SNPs at HBS1L-MYB intergenic region which were shown to modify HbF levels. The different genotypes can be determined based on the position and the fluorescent colour of the peaks in a single electropherogram. DNA sequencing and restriction fragment length polymorphism (PCR-RFLP) assays were used to verify genotyping results obtained by SNaPshot minisequencing. CONCLUSIONS In summary, we propose two multiplex assays based on the SNaPshot minisequencing approach for the simultaneous identification of SNPs located at the BCL11A gene and HBS1L-MYB intergenic region which have an effect on HbF levels. The assays can be easily applied for accurate, time and cost efficient genotyping of the selected SNPs in various populations.
Collapse
Affiliation(s)
| | | | | | - Marina Kleanthous
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Agios Dometios, Nicosia 1683, Cyprus.
| |
Collapse
|
48
|
Musallam KM, Rivella S, Vichinsky E, Rachmilewitz EA. Non-transfusion-dependent thalassemias. Haematologica 2013; 98:833-44. [PMID: 23729725 DOI: 10.3324/haematol.2012.066845] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Non-transfusion-dependent thalassemias include a variety of phenotypes that, unlike patients with beta (β)-thalassemia major, do not require regular transfusion therapy for survival. The most commonly investigated forms are β-thalassemia intermedia, hemoglobin E/β-thalassemia, and α-thalassemia intermedia (hemoglobin H disease). However, transfusion-independence in such patients is not without side effects. Ineffective erythropoiesis and peripheral hemolysis, the hallmarks of disease process, lead to a variety of subsequent pathophysiologies including iron overload and hypercoagulability that ultimately lead to a number of serious clinical morbidities. Thus, prompt and accurate diagnosis of non-transfusion-dependent thalassemia is essential to ensure early intervention. Although several management options are currently available, the need to develop more novel therapeutics is justified by recent advances in our understanding of the mechanisms of disease. Such efforts require wide international collaboration, especially since non-transfusion-dependent thalassemias are no longer bound to low- and middle-income countries but have spread to large multiethnic cities in Europe and the Americas due to continued migration.
Collapse
Affiliation(s)
- Khaled M Musallam
- Department of Medicine and Medical Specialties, IRCCS Ca’ Granda Foundation Maggiore Policlinico Hospital, University of Milan, Milan, Italy.
| | | | | | | |
Collapse
|
49
|
Toma S, Tenorio M, Oakley M, Thein SL, Clark BE. Two Novel Mutations (HBG1: c.-250C>T andHBG2: c.-250C>T) Associated With Hereditary Persistence of Fetal Hemoglobin. Hemoglobin 2013; 38:67-9. [DOI: 10.3109/03630269.2013.848365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Banan M, Bayat H, Namdar-Aligoodarzi P, Azarkeivan A, Kamali K, Daneshmand P, Zaker-Kandjani B, Najmabadi H. Utility of the multivariate approach in predicting β-thalassemia intermedia or β-thalassemia major types In Iranian patients. Hemoglobin 2013; 37:413-22. [PMID: 23805990 DOI: 10.3109/03630269.2013.805418] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recently, five genetic modifiers [β-globin mutations, coinheritance of α-thalassemia (α-thal), XmnI polymorphism and single nucleotide polymorphisms (SNPs) in the BCL11A and HBS1L-MYB loci] were used to predict the β-thal major (β-TM) or β-thal intermedia (β-TI) types in 106 French patients with 83.2% accuracy. The dichotomous grouping was based on the age when the patient received his/her first transfusion (4 years). Here, a similar study was conducted in a cohort of 306 Iranian β-thal patients having distinct β-globin mutations and minor allele frequencies of key SNPs in these loci. Multivariate regression analyses and a simple scoring system were used to predict the β-TM/β-TI types using three scenarios: 1) when considering only the severe β-TM and the mild β-TI cases, 2) using clinical parameters for β-thal typing, and 3) using age at first transfusion as the basis for classification. Using these scenarios, the β-thal types could be correctly predicted in 77.6, 75.5 and 68.0% of cases, respectively.
Collapse
Affiliation(s)
- Mehdi Banan
- Genetics Research Centre, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | |
Collapse
|