51
|
Papadopoulos P, Kafasi A, De Cuyper IM, Barroca V, Lewandowski D, Kadri Z, Veldthuis M, Berghuis J, Gillemans N, Benavente Cuesta CM, Grosveld FG, van Zwieten R, Philipsen S, Vernet M, Gutiérrez L, Patrinos GP. Mild dyserythropoiesis and β-like globin gene expression imbalance due to the loss of histone chaperone ASF1B. Hum Genomics 2020; 14:39. [PMID: 33066815 PMCID: PMC7566067 DOI: 10.1186/s40246-020-00283-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/10/2020] [Indexed: 01/09/2023] Open
Abstract
The expression of the human β-like globin genes follows a well-orchestrated developmental pattern, undergoing two essential switches, the first one during the first weeks of gestation (ε to γ), and the second one during the perinatal period (γ to β). The γ- to β-globin gene switching mechanism includes suppression of fetal (γ-globin, HbF) and activation of adult (β-globin, HbA) globin gene transcription. In hereditary persistence of fetal hemoglobin (HPFH), the γ-globin suppression mechanism is impaired leaving these individuals with unusual elevated levels of fetal hemoglobin (HbF) in adulthood. Recently, the transcription factors KLF1 and BCL11A have been established as master regulators of the γ- to β-globin switch. Previously, a genomic variant in the KLF1 gene, identified by linkage analysis performed on twenty-seven members of a Maltese family, was found to be associated with HPFH. However, variation in the levels of HbF among family members, and those from other reported families carrying genetic variants in KLF1, suggests additional contributors to globin switching. ASF1B was downregulated in the family members with HPFH. Here, we investigate the role of ASF1B in γ- to β-globin switching and erythropoiesis in vivo. Mouse-human interspecies ASF1B protein identity is 91.6%. By means of knockdown functional assays in human primary erythroid cultures and analysis of the erythroid lineage in Asf1b knockout mice, we provide evidence that ASF1B is a novel contributor to steady-state erythroid differentiation, and while its loss affects the balance of globin expression, it has no major role in hemoglobin switching.
Collapse
Affiliation(s)
- Petros Papadopoulos
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.
| | - Athanassia Kafasi
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, AMC, UvA, Amsterdam, The Netherlands
| | - Iris M De Cuyper
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, AMC, UvA, Amsterdam, The Netherlands
| | - Vilma Barroca
- UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris and Université de Paris-Saclay, CEA, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
- U1274, Inserm, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
| | - Daniel Lewandowski
- UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris and Université de Paris-Saclay, CEA, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
- U1274, Inserm, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
| | - Zahra Kadri
- Division of Innovative Therapies, UMR1184, Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Martijn Veldthuis
- Laboratory of Red Blood Cell Diagnostics, Sanquin Diagnostics, Amsterdam, The Netherlands
| | - Jeffrey Berghuis
- Laboratory of Red Blood Cell Diagnostics, Sanquin Diagnostics, Amsterdam, The Netherlands
| | - Nynke Gillemans
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Celina María Benavente Cuesta
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Rob van Zwieten
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, AMC, UvA, Amsterdam, The Netherlands
- Laboratory of Red Blood Cell Diagnostics, Sanquin Diagnostics, Amsterdam, The Netherlands
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Muriel Vernet
- UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris and Université de Paris-Saclay, CEA, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
| | - Laura Gutiérrez
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, AMC, UvA, Amsterdam, The Netherlands
- Platelet Research Lab -Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)-, Department of Medicine -University of Oviedo-, Oviedo, Spain
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, University of Patras School of Health Sciences, Patras, Greece
- Department of Pathology, College of Medicine and Health Sciences and Zayed Center of Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
52
|
Kulczynska-Figurny K, Bieker JJ, Siatecka M. Severe anemia caused by dominant mutations in Krüppel-like factor 1 (KLF1). MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 786:108336. [PMID: 33339573 DOI: 10.1016/j.mrrev.2020.108336] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/23/2020] [Accepted: 09/29/2020] [Indexed: 10/23/2022]
Abstract
The etiology and severity of anemia, a common blood disorder, are diverse. Dominant mutations in Krüppel-like factor 1 (KLF1/EKLF) underlie the molecular basis for some of them. KLF1 is a zinc finger transcription factor that plays an essential role in red blood cell proliferation and differentiation. Mutations have been identified in the KLF1 gene that cause hematologic diseases. Two of these alter one allele but generate an extreme phenotype: the mouse Nan mutation (E339D) leads to hemolytic neonatal anemia with hereditary spherocytosis, and the human CDA mutation (E325K) causes congenital dyserythropoietic anemia (CDA) type IV. These modify functionally important amino acids in the zinc finger DNA-binding domain at positions involved in direct interactions with regulatory elements of KLF1's target genes. Although the two dominant mutations alter the same evolutionarily conserved glutamic acid residue, the substitutions are not equivalent and lead to divergent consequences for the molecular mechanisms underlying activity of these mutants, particularly in recognition and interaction with their unique binding sites. Consequently, the properties of the protein are transformed such that it acquires novel dominant characteristics whose effects may not be limited to the erythroid compartment. KLF1 mutants cause loss-of-function/haploinsufficiency effects on some KLF1 wild-type target genes, while at the same time gain-of-function effects activate ectopic sites and neomorphic gene expression. Such anomalies not only lead to intrinsic red cell problems, but also to expression of non-erythroid genes that systemically disturb organ development. This review highlights recent molecular, biochemical, and genetic studies of KLF1 mutants, particularly the dramatic consequences that come from just a single amino acid change. The study of these variants provides an important contribution to the overall understanding of the DNA-protein interface of the zinc finger subtype of transcription factors, and the potential clinical consequences of what might appear to be a minor change in sequence.
Collapse
Affiliation(s)
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miroslawa Siatecka
- Department of Genetics, Faculty of Biology, University of Adam Mickiewicz, Poznan, 61-614, Poland.
| |
Collapse
|
53
|
Tepakhan W, Kanjanaopas S, Srewaradachpisal K. Association Between Genetic Polymorphisms and Hb F Levels in Heterozygous β-Thalassemia 3.5 kb Deletions. Hemoglobin 2020; 44:338-343. [PMID: 32878504 DOI: 10.1080/03630269.2020.1811117] [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: 10/23/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in several genetic modifying factors have been related to Hb F levels, including Gγ XmnI polymorphism, B-cell lymphoma/leukemia 11 A (BCL11A), HBS1L-MYB intergenic polymorphism (HMIP) and a mutation in the Krüppel-like factor 1 (KLF1). This study aimed to determine whether genetic variability of these modifying factors affects Hb F levels in heterozygous β-thalassemia (β-thal) 3.5 kb deletion (NC_000011.10: g.5224302-5227791del13490bp). A total of 111 β-thal 3.5 kb deletion carriers with Hb F levels ranging from 0.9 to 18.4% was recruited for this study. Genotyping of SNPs including HBG2 rs7482144, HMIP rs4895441 and rs9399137, BCL11A rs4671393 and KLF1 rs2072596 was identified. Multiple regression analyses showed that only two SNPs (HMIP rs4895441 and rs9399137) influenced Hb F levels. Interestingly, a combination of these two SNPs was associated with higher Hb F levels. Our study is the first to demonstrate that the rs4895441, rs9399137 of HMIP are associated with elevated Hb F levels in the heterozygous β-thal 3.5 kb deletion.
Collapse
Affiliation(s)
- Wanicha Tepakhan
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Sataron Kanjanaopas
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Korntip Srewaradachpisal
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
54
|
Mansoor A, Mansoor MO, Patel JL, Zhao S, Natkunam Y, Bieker JJ. KLF1/EKLF expression in acute leukemia is correlated with chromosomal abnormalities. Blood Cells Mol Dis 2020; 83:102434. [PMID: 32311573 DOI: 10.1016/j.bcmd.2020.102434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022]
Abstract
KLF1 (EKLF) is a master regulator of erythropoiesis and controls expression of a wide array of target genes. We interrogated human tissue microarray samples via immunohistological analysis to address whether levels of KLF1 protein are associated with leukemia. We have made the unexpected findings that higher KLF1 levels are correlated with cells containing abnormal chromosomes, and that high KLF1 expression is not limited to acute myeloid leukemia (AML) associated with erythroid/megakaryoblastic differentiation. Expression of KLF1 is associated with poor survival. Further analyses reveal that KLF1 directly regulates a number of genes that play a role in chromosomal integrity. Together these results suggest that monitoring KLF1 levels may provide a new marker for risk stratification and prognosis in patients with AML.
Collapse
Affiliation(s)
- Adnan Mansoor
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Mohammad Omer Mansoor
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Jay L Patel
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - James J Bieker
- Department of Cell, Developmental, & Regenerative Biology, Black Family Stem Cell Institute, Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA.
| |
Collapse
|
55
|
Tatu T. Wide range of F cell levels in healthy Thai adults: Influence of Swiss-type hereditary persistence of foetal haemoglobin & β-haemoglobinopathy. Indian J Med Res 2020; 150:161-166. [PMID: 31670271 PMCID: PMC6829780 DOI: 10.4103/ijmr.ijmr_1954_17] [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: 11/04/2022] Open
Abstract
Background & objectives: Swiss-type hereditary persistence of foetal haemoglobin (HPFH) has been shown to be responsible for the wide range of F cell levels in healthy Thai adults. However, a survey for F cells in healthy Thai adults has not been performed. This study was conducted to determine the F cell distribution in adult Thai blood donors and to assess the possible involvement of β-thalassaemia and haemoglobin E (HbE) carriers in increased HbF levels. Methods: Thai blood donors (n=375, 205 males and 170 females) were included in the study. Blood samples were collected for measuring haemoglobin (Hb) concentration and haematocrit (Hct) and F cell levels. Hb and Hct levels were determined by automated blood counter, while F cells were quantified by flow cytometric analysis of F cells stained by fluorescein isothiocyanate-conjugated anti γ-globin monoclonal antibody. Finally, F cell levels were compared between blood samples having mean corpuscular volume (MCV) <80 fl and ≥80 fl as well as between β-haemoglobinopathies (HbE and β-thalassaemia carriers) and normal adults. Results: F cell levels varied markedly spanning 0.80-39.2 per cent with a positively skewed distribution. Thirty two per cent of these individuals had F cell levels more than the 4.5 per cent cut-off point. F cell levels in females were significantly higher than those in males (P<0.05). F cell levels in individuals having MCV <80 fl were significantly higher than those having MCV ≥80 fl (P<0.05). β-haemoglobinopathy (HbE and β-thalassaemia carriers) had significantly higher F cell levels than normal individuals (P<0.05). Interpretation & conclusions: The present results showed that besides Swiss-type HPFH, the β-haemoglobinopathy was expected to be involved in increased F cell levels in adult Thais. Thus, influence of β-haemoglobinopathy must be considered in interpreting F cell levels in area endemic of this globin disorder.
Collapse
Affiliation(s)
- Thanusak Tatu
- Department of Medical Technology, Division of Clinical Microscopy, Research Center for Hematology & Health Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
56
|
A Krüppel-like factor 1 ( KLF1) Mutation Associated with Severe Congenital Dyserythropoietic Anemia Alters Its DNA-Binding Specificity. Mol Cell Biol 2020; 40:MCB.00444-19. [PMID: 31818881 DOI: 10.1128/mcb.00444-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/04/2019] [Indexed: 11/20/2022] Open
Abstract
Krüppel-like factor 1 (KLF1/EKLF) is a transcription factor that globally activates genes involved in erythroid cell development. Various mutations are identified in the human KLF1 gene. The E325K mutation causes congenital dyserythropoietic anemia (CDA) type IV, characterized by severe anemia and non-erythroid-cell-related symptoms. The CDA mutation is in the second zinc finger of KLF1 at a position functionally involved in its interactions with DNA. The molecular parameters of how CDA-KLF1 exerts its biological effects have not been addressed. Here, using an in vitro selection strategy, we determined the preferred DNA-binding site for CDA-KLF1. Binding to the deduced consensus sequence is supported by in vitro gel shifts and by in vivo functional reporter gene studies. Two significant changes compared to wild-type (WT) binding are observed: G is selected as the middle nucleotide, and the 3' portion of the consensus sequence is more degenerate. As a consequence, CDA-KLF1 did not bind the WT consensus sequence. However, activation of ectopic sites is promoted. Continuous activation of WT target genes occurs if they fortuitously contain the novel CDA site nearby. Our findings provide a molecular understanding of how a single mutation in the KLF1 zinc finger exerts effects on erythroid physiology in CDA type IV.
Collapse
|
57
|
Reactivation of γ-globin expression through Cas9 or base editor to treat β-hemoglobinopathies. Cell Res 2020; 30:276-278. [PMID: 31911671 DOI: 10.1038/s41422-019-0267-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
|
58
|
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
|
59
|
Jiang H, Jiang F, Li J, Tang F, Li DZ. Congenital Nonspherocytic Hemolytic Anemia Caused by Krüppel-Like Factor 1 Gene Variants: Another Case Report. Hemoglobin 2019; 43:292-295. [PMID: 31645145 DOI: 10.1080/03630269.2019.1680384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this study, we report on a compound heterozygote for variants in the key erythroid transcription factor Krüppel-like factor 1 (KLF1) gene in a patient who presented with severe, transfusion-dependent hemolytic anemia. The red cells were normochromic and normocytic, and resembled those seen in patients with congenital nonspherocytic hemolytic anemia (CNSHA). Next generation sequencing (NGS) revealed that the patient was a compound heterozygote for the KLF1 frameshift variant c.519_525dup (p.Gly176ArgfsTer179) and a missense variant c.1012C>A (p.Pro338Thr). This report adds to the wide clinical spectrum of KLF1 gene variants. We suggest that loss of KLF1 should be considered in otherwise unexplained cases of congenital hemolytic anemia.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Pediatric Hematology-Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Fan Jiang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jian Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Fang Tang
- Neonatal Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Dong-Zhi Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
60
|
Zhang L, Zhang Q, Tang Y, Cong P, Ye Y, Chen S, Zhang X, Chen Y, Zhu B, Cai W, Chen S, Cai R, Guo X, Zhang C, Zhou Y, Zou J, Liu Y, Chen B, Yan S, Chen Y, Zhou Y, Ding H, Li X, Chen D, Zhong J, Shang X, Liu X, Qi M, Xu X. LOVD-DASH: A comprehensive LOVD database coupled with diagnosis and an at-risk assessment system for hemoglobinopathies. Hum Mutat 2019; 40:2221-2229. [PMID: 31286593 PMCID: PMC6899610 DOI: 10.1002/humu.23863] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
Hemoglobinopathies are the most common monogenic disorders worldwide. Substantial effort has been made to establish databases to record complete mutation spectra causing or modifying this group of diseases. We present a variant database which couples an online auxiliary diagnosis and at-risk assessment system for hemoglobinopathies (DASH). The database was integrated into the Leiden Open Variation Database (LOVD), in which we included all reported variants focusing on a Chinese population by literature peer review-curation and existing databases, such as HbVar and IthaGenes. In addition, comprehensive mutation data generated by high-throughput sequencing of 2,087 hemoglobinopathy patients and 20,222 general individuals from southern China were also incorporated into the database. These sequencing data enabled us to observe disease-causing and modifier variants responsible for hemoglobinopathies in bulk. Currently, 371 unique variants have been recorded; 265 of 371 were described as disease-causing variants, whereas 106 were defined as modifier variants, including 34 functional variants identified by a quantitative trait association study of this high-throughput sequencing data. Due to the availability of a comprehensive phenotype-genotype data set, DASH has been established to automatically provide accurate suggestions on diagnosis and genetic counseling of hemoglobinopathies. LOVD-DASH will inspire us to deal with clinical genotyping and molecular screening for other Mendelian disorders.
Collapse
Affiliation(s)
- Li Zhang
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Qianqian Zhang
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | | | - Peikuan Cong
- Institute for Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuhua Ye
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Shiping Chen
- BGI Clinical Laboratories-Shenzhen, Shenzhen, Guangdong, China
| | - Xinhua Zhang
- Department of Hematopathology, 303rd Hospital of the People's Liberation Army, Nanning, Guangxi, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Baosheng Zhu
- Genetic Diagnosis Center, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Wangwei Cai
- School of Basic Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Shaoke Chen
- Department of Pediatrics, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, Guangxi, China
| | - Ren Cai
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Xiaoling Guo
- Department of Pediatrics, Maternity and Child Health Care Hospital of Foshan City, Foshan, Guangdong, China
| | - Chonglin Zhang
- Department of Clinical Laboratory, Guilin Women and Children health care hospital, Guilin, Guangxi, China
| | - Yuqiu Zhou
- Department of Clinical Laboratory, Zhuhai Municipal Maternity and Child Healthcare Hospital, Zhuhai, Guangdong, China
| | - Jie Zou
- Department of Clinical Laboratory, Maternal and Child Health Hospital in Meizhou, Meizhou, Guangdong, China
| | - Yanhui Liu
- Department of Prenatal Diagnosis Center, Maternal and Child Health Hospital, Dongguan, Guangdong, China
| | - Biyan Chen
- Department of Clinical Laboratory, Baise Women and Children Care Hospital, Baise, Guangxi, China
| | - Shanhuo Yan
- Department of Clinical Laboratory, Qinzhou Maternal and Child Health Hospital, Qinzhou, Guangxi, China
| | - Yajun Chen
- Center For Prenatal Diagnosis Shaoguan, Shaoguan Municipal Maternity and Child Healthcare Hospital, Guangdong, China
| | - Yuehong Zhou
- Department of Clinical Laboratory, The People's Hospital of Yunfu City, Yunfu, Guangdong, China
| | - Hongmei Ding
- Department of Clinical Laboratory, Pingguo Women and Children Care Hospital, Baise, Guangxi, China
| | - Xiarong Li
- Beijing GeneDock Technology Company, Beijing, China
| | - Dianyu Chen
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmei Zhong
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Xuan Shang
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Xuanzhu Liu
- Beijing GeneDock Technology Company, Beijing, China
| | - Ming Qi
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Center for Genetic & Genomic Medicine, JamesWatson Institute of Genome Sciences, Zhejiang University Medical School 1st Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Xiangmin Xu
- Department of Medical Genetics, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| |
Collapse
|
61
|
Nicolau M, Vargas S, Silva M, Coelho A, Ferreira E, Mendonça J, Vieira L, Kjöllerström P, Maia R, Silva R, Dias A, Ferreira T, Morais A, Soares IM, Lavinha J, Faustino P. Genetic modulators of fetal hemoglobin expression and ischemic stroke occurrence in African descendant children with sickle cell anemia. Ann Hematol 2019; 98:2673-2681. [PMID: 31478061 DOI: 10.1007/s00277-019-03783-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/11/2019] [Indexed: 02/06/2023]
Abstract
Sickle cell anemia (SCA) is an autosomal recessive monogenic disease with significant clinical variability. Cerebrovascular disease, particularly ischemic stroke, is one of the most severe complications of SCA in children. This study aimed to investigate the influence of genetic variants on the levels of fetal hemoglobin (Hb F) and biochemical parameters related with chronic hemolysis, as well as on ischemic stroke risk, in ninety-one unrelated SCA patients, children of sub-Saharan progenitors. Our results show that a higher Hb F level has an inverse relationship with the occurrence of stroke, since the group of patients who suffered stroke presents a significantly lower mean Hb F level (5.34 ± 4.57% versus 9.36 ± 6.48%; p = 0.024). Furthermore, the co-inheritance of alpha-thalassemia improves the chronic hemolytic pattern, evidenced by a decreased reticulocyte count (8.61 ± 3.58% versus 12.85 ± 4.71%; p < 0.001). In addition, our findings have confirmed the importance of HBG2 and BCL11A loci in the regulation of Hb F expression in sub-Saharan African SCA patients, as rs7482144_A, rs11886868_C, and rs4671393_A alleles are significantly associated with a considerable increase in Hb F levels (p = 0.019, p = 0.026, and p = 0.028, respectively). Concerning KLF1, twelve different variants were identified, two of them novel. Seventy-three patients (80.2%) presented at least one variant in this gene. However, no correlation was observed between the presence of these variants and Hb F level, severity of hemolysis, or stroke occurrence, which is consistent with their in silico-predicted minor functional consequences. Thus, we conclude that the prevalence of functional KLF1 variants in a sub-Saharan African background does not seem to be relevant to SCA clinical modulation.
Collapse
Affiliation(s)
- Marta Nicolau
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Sofia Vargas
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Andreia Coelho
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Emanuel Ferreira
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Joana Mendonça
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Luís Vieira
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal.,ToxOmics, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Paula Kjöllerström
- Unidade de Hematologia, Hospital de Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central (CHULC), Lisbon, Portugal
| | - Raquel Maia
- Unidade de Hematologia, Hospital de Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central (CHULC), Lisbon, Portugal
| | - Rita Silva
- Unidade de Neuropediatria, Hospital de Dona Estefânia, CHULC, Lisbon, Portugal
| | - Alexandra Dias
- Núcleo de Hematologia, Departamento de Pediatria, Hospital Prof. Doutor Fernando Fonseca, Amadora, Portugal
| | - Teresa Ferreira
- Núcleo de Hematologia, Departamento de Pediatria, Hospital Prof. Doutor Fernando Fonseca, Amadora, Portugal
| | - Anabela Morais
- Departamento de Pediatria, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | | | - João Lavinha
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal.,BioISI, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal. .,Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal. .,Unidade de Investigação e Desenvolvimento, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Avenida Padre Cruz, 1649-016, Lisbon, Portugal.
| |
Collapse
|
62
|
Barrera-Reyes PK, Tejero ME. Genetic variation influencing hemoglobin levels and risk for anemia across populations. Ann N Y Acad Sci 2019; 1450:32-46. [PMID: 31385320 DOI: 10.1111/nyas.14200] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/30/2019] [Accepted: 07/05/2019] [Indexed: 01/19/2023]
Abstract
Hemoglobin (Hb) concentration is the outcome of the interaction between genetic variation and environmental factors, including nutritional status, sex, age, and altitude. Genetic diversity influencing this protein is complex and varies widely across populations. Variants related to abnormal Hb or altered characteristics of the erythrocytes increase the risk for anemia. The most prevalent are related to the inherited globin abnormalities affecting Hb production and structure. Malaria-endemic regions harbor the highest frequencies of variants associated with the most frequent monogenic diseases and the risk for nonnutritional anemia and are considered as public health problems. Variation in genes encoding for enzymes and membrane proteins in red blood cells also influence erythrocyte life span and risk for anemia. Most of these variants are rare. Interindividual variability of hematological parameters is also influenced by common genetic variation across the whole genome. Some of the identified variants are associated with Hb production, erythropoiesis, and iron metabolism. Specialized databases have been developed to organize and update the large body of available information on genetic variation related to Hb variation, their frequency, geographical distribution, and clinical significance. Our present review analyzed the underlying genetic factors that affect Hb concentrations, their clinical relevance, and geographical distribution across populations.
Collapse
Affiliation(s)
- Paloma K Barrera-Reyes
- Laboratorio de Nutrigenómica y Nutrigenética, Instituto Nacional de Medicina Genómica, Ciudad de, México, Mexico.,Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de, México, Mexico
| | - M Elizabeth Tejero
- Laboratorio de Nutrigenómica y Nutrigenética, Instituto Nacional de Medicina Genómica, Ciudad de, México, Mexico
| |
Collapse
|
63
|
Fanis P, Kousiappa I, Phylactides M, Kyrri A, Hadjigavriel M, Christou S, Sitarou M, Kleanthous M. A novel mutation in the erythroid transcription factor KLF1 is likely responsible for ameliorating β-thalassemia major. Hum Mutat 2019; 40:1768-1780. [PMID: 31115947 PMCID: PMC6790707 DOI: 10.1002/humu.23817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/02/2019] [Accepted: 05/05/2019] [Indexed: 12/02/2022]
Abstract
We describe the identification of a novel missense mutation in the second zinc finger of KLF1 in two siblings who, based on their genotype, are predicted to suffer from beta thalassemia major but are, in fact, transfusion‐free and in good health. These individuals, as well as two additional members of the same family also carrying this KLF1 mutation, exhibit high levels of fetal hemoglobin (HbF). KLF1 is an erythroid transcription factor, which plays a critical role in the regulation of the developmental switch between fetal and adult hemoglobin by regulating the expression of a multitude of genes including that of BCL11A. The mutation appears to be the main candidate responsible for the beta thalassemia‐ameliorating effect as this segregates with the observed phenotype and also exogenous expression of the KLF1 mutant protein in human erythroid progenitor cells resulted in the induction of γ‐globin, without, however, affecting BCL11A levels. This report adds to the weight of evidence that heterozygous KLF1 mutations can ameliorate the severity of the β‐thalassemia major phenotype.
Collapse
Affiliation(s)
- Pavlos Fanis
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Ioanna Kousiappa
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marios Phylactides
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Andreani Kyrri
- Population Screening Laboratory, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | | | | | - Maria Sitarou
- Thalassaemia Clinic, Larnaca General Hospital, Larnaca, Cyprus
| | - Marina Kleanthous
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
64
|
Yang K, Ren Q, Wu Y, Zhou Y, Yin X. A Case of Hereditary Spherocytosis Caused by a Novel Homozygous Mutation in the SPTB Gene Misdiagnosed as β-Thalassemia Intermedia Due to a KLF1 Gene Mutation. Hemoglobin 2019; 43:140-144. [PMID: 31190573 DOI: 10.1080/03630269.2019.1620764] [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: 10/26/2022]
Abstract
We report a rare case of hereditary spherocytosis (HS) and hereditary persistence of fetal hemoglobin (Hb) (HPFH) complicated with a β-thalassemia (β-thal) trait and a Krüppel-like factor 1 (KLF1) gene mutation misdiagnosed as β-thal intermedia (β-TI) due to a high percentage of Hb F. The proband presented with pale skin, jaundice and splenomegaly. Analysis of the thalassemia gene indicated βcodon 17/βA (HBB: c.52A>T), while Hb analysis showed significantly increased Hb F levels. The proband was diagnosed to carry β-TI, and a blood transfusion regimen together with iron chelation treatment was recommended. Due to the difference between the phenotype and genotype, next generation sequencing (NGS) was performed and the proband was found to carry a homozygous mutation on the SPTB gene combined with a heterozygous mutation in KLF1. An eosin-5-maleimide binding test (EMA-BT) showed that the mean fluorescence intensity decreased by 47.1%. The proband was finally diagnosed with HS and HPFH complicated with a β-thal trait and the high percentage of Hb F was believed to be ascribed to the KLF1 gene mutation, which is frequent in areas where thalassemia is prevalent. For patients with a β gene mutation accompanying significantly high percentage of Hb F, the diagnosis of β-TI could be warranted, and the influence of the KLF1 gene mutation should be carefully excluded to avoid misdiagnosis of other types of hereditary hemolytic diseases.
Collapse
Affiliation(s)
- Kun Yang
- a Graduate School of Guangxi University of Chinese Medicine , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China.,b Department of Hematology , The 923rd Hospital of the Peoples Liberation Army , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China
| | - Quan Ren
- b Department of Hematology , The 923rd Hospital of the Peoples Liberation Army , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China.,c Graduate School of Guilin Medical University , Guilin , Guangxi Zhuang Autonomous Region , People's Republic of China
| | - Yi Wu
- b Department of Hematology , The 923rd Hospital of the Peoples Liberation Army , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China.,c Graduate School of Guilin Medical University , Guilin , Guangxi Zhuang Autonomous Region , People's Republic of China
| | - Yali Zhou
- b Department of Hematology , The 923rd Hospital of the Peoples Liberation Army , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China
| | - Xiaolin Yin
- b Department of Hematology , The 923rd Hospital of the Peoples Liberation Army , Nanning , Guangxi Zhuang Autonomous Region , People's Republic of China
| |
Collapse
|
65
|
Ilsley MD, Huang S, Magor GW, Landsberg MJ, Gillinder KR, Perkins AC. Corrupted DNA-binding specificity and ectopic transcription underpin dominant neomorphic mutations in KLF/SP transcription factors. BMC Genomics 2019; 20:417. [PMID: 31126231 PMCID: PMC6534859 DOI: 10.1186/s12864-019-5805-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background Mutations in the transcription factor, KLF1, are common within certain populations of the world. Heterozygous missense mutations in KLF1 mostly lead to benign phenotypes, but a heterozygous mutation in a DNA-binding residue (E325K in human) results in severe Congenital Dyserythropoietic Anemia type IV (CDA IV); i.e. an autosomal-dominant disorder characterized by neonatal hemolysis. Results To investigate the biochemical and genetic mechanism of CDA IV, we generated murine erythroid cell lines that harbor tamoxifen-inducible (ER™) versions of wild type and mutant KLF1 on a Klf1−/− genetic background. Nuclear translocation of wild type KLF1 results in terminal erythroid differentiation, whereas mutant KLF1 results in hemolysis without differentiation. The E to K variant binds poorly to the canonical 9 bp recognition motif (NGG-GYG-KGG) genome-wide but binds at high affinity to a corrupted motif (NGG-GRG-KGG). We confirmed altered DNA-binding specificity by quantitative in vitro binding assays of recombinant zinc-finger domains. Our results are consistent with previously reported structural data of KLF-DNA interactions. We employed 4sU-RNA-seq to show that a corrupted transcriptome is a direct consequence of aberrant DNA binding. Conclusions Since all KLF/SP family proteins bind DNA in an identical fashion, these results are likely to be generally applicable to mutations in all family members. Importantly, they explain how certain mutations in the DNA-binding domain of transcription factors can generate neomorphic functions that result in autosomal dominant disease. Electronic supplementary material The online version of this article (10.1186/s12864-019-5805-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa D Ilsley
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia.,School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Stephen Huang
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia.,School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Graham W Magor
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Kevin R Gillinder
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia. .,Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia.
| | - Andrew C Perkins
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
66
|
Xie XM, Liu YN, Li J, Jiang F, Li DZ. A Krüppel-Like Factor 1 Gene Mutation Ameliorates the Severity of β-Thalassemia: A Case Report. Hemoglobin 2019; 43:137-139. [PMID: 31111750 DOI: 10.1080/03630269.2019.1607373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Patients with the β0/β0 type of β-thalassemia (β-thal) usually present as β-thal major (β-TM), and are transfusion-dependent. However, the clinical and hematological features of β-thal can be modulated by different modifiers, resulting in a wide range of clinical severity even in patients with the same genotypes. We report a Chinese family with twin brothers, both of whom had the same genotype of β0/β0. One twin was diagnosed as β-TM at 4 months of age and had regularly been transfused; conversely the other twin with a KLF1 (Krüppel-like factor 1) gene mutation, behaved as β-thal intermedia (β-TI), and had never been transfused. Our findings indicate that KLF1 mutations have a role in modulating the phenotypic severity of β-thal. The exact investigation of KLF1 modifiers is necessary in areas where globin gene disorders are most prevalent. This will be helpful in genetic counseling and optimizing the guidelines for prenatal diagnosis (PND) programs.
Collapse
Affiliation(s)
- Xing-Mei Xie
- a Prenatal Diagnostic Center , Guangzhou Women and Children's Medical Center affiliated to Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Ying-Na Liu
- b Department of Obstetrics and Gynecology , Shenzhen Hospital of Southern Medical University , Shenzhen , Guangdong Province , People's Republic of China
| | - Jian Li
- a Prenatal Diagnostic Center , Guangzhou Women and Children's Medical Center affiliated to Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Fan Jiang
- a Prenatal Diagnostic Center , Guangzhou Women and Children's Medical Center affiliated to Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Dong-Zhi Li
- a Prenatal Diagnostic Center , Guangzhou Women and Children's Medical Center affiliated to Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| |
Collapse
|
67
|
Zhang J, Yang Y, Li P, Yan Y, Lv T, Zhao T, Zeng X, Li D, Zhou X, Chen H, Su J, Yang T, He J, Zhu B. Analysis of deletional hereditary persistence of fetal hemoglobin/δβ-thalassemia and δ-globin gene mutations in Southerwestern China. Mol Genet Genomic Med 2019; 7:e706. [PMID: 31044540 PMCID: PMC6565566 DOI: 10.1002/mgg3.706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 01/07/2023] Open
Abstract
Background Deletional hereditary persistence of fetal hemoglobin (HPFH)/δβ‐thalassemia and δ‐thalassemia are rare inherited disorders which may complicate the diagnosis of β‐thalassemia. The aim of this study was to reveal the frequency of these two disorders in Southwestern China. Methods A total of 33,596 subjects were enrolled for deletional HPFH/δβ‐thalassemia, and positive individuals with high fetal hemoglobin (Hb F) level were diagnosed by multiplex ligation‐dependent probe amplification (MLPA). A total of 17,834 subjects were analyzed for mutations in the δ‐globin gene. Positive samples with low Hb A2 levels were confirmed by δ‐globin gene sequencing. Furthermore, the pathogenicity and construction of a selected δ‐globin mutation were analyzed. Results A total of 92 suspected cases with Hb F ≥5.0% were further characterized by MLPA. Eight different deletional HPFH/δβ‐thalassemia were observed at a frequency of 0.024%. In addition, 195 cases suspected to have a δ‐globin gene mutation (Hb A2 ≤2.0%) were characterized by molecular analysis. δ‐Globin gene mutation was found at a frequency of 0.49% in Yunnan. The pathogenicity and construction for a selected δ‐globin mutation was predicted. Conclusion Screening of these two disorders was analyzed in Southwestern China, which could define the molecular basis of these conditions in this population.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Obstetrics and GynecologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
- Department of HematologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Yang Yang
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Peng Li
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Yuanlong Yan
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tao Lv
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tingting Zhao
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Xiaohong Zeng
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Dongmei Li
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Xiaoyan Zhou
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Hong Chen
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Jie Su
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tonghua Yang
- Department of HematologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Jing He
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Baosheng Zhu
- Department of Obstetrics and GynecologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| |
Collapse
|
68
|
Genetic Variants Within the Erythroid Transcription Factor, KLF1, and Reduction of the Expression of Lutheran and Other Blood Group Antigens: Review of the In(Lu) Phenotype. Transfus Med Rev 2019; 33:111-117. [DOI: 10.1016/j.tmrv.2019.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 11/22/2022]
|
69
|
Varricchio L, Planutis A, Manwani D, Jaffray J, Mitchell WB, Migliaccio AR, Bieker JJ. Genetic disarray follows mutant KLF1-E325K expression in a congenital dyserythropoietic anemia patient. Haematologica 2019; 104:2372-2380. [PMID: 30872368 PMCID: PMC6959163 DOI: 10.3324/haematol.2018.209858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Congenital dyserythropoietic anemia type IV is caused by a heterozygous mutation, Glu325Lys (E325K), in the KLF1 transcription factor. Molecular characteristics of this disease have not been clarified, partly due to its rarity. We expanded erythroid cells from a patient's peripheral blood and analyzed its global expression pattern. We find that a large number of erythroid pathways are disrupted, particularly those related to membrane transport, globin regulation, and iron utilization. The altered genetics lead to significant deficits in differentiation. Glu325 is within the KLF1 zinc finger domain at an amino acid critical for site specific DNA binding. The change to Lys is predicted to significantly alter the target site recognition sequence, both by subverting normal recognition and by enabling interaction with novel sites. Consistent with this, we find high level ectopic expression of genes not normally present in the red cell. These altered properties explain patients' clinical and phenotypic features, and elucidate the dominant character of the mutation.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antanas Planutis
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepa Manwani
- Division of Hematology/Oncology, The Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Julie Jaffray
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - W Beau Mitchell
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Dipartimento di Scienze Biomediche e NeuroMotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - James J Bieker
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA .,Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
70
|
Tamaddoni A, Khabaz Astaneh S, Tabaripour R, Akhavan-Niaki H. Krüppel-Like Factor 1 Gene Mutations in Thalassemia Patients from North Iran: Report of a New Mutation Associated with β-Thalassemia Intermedia. Hemoglobin 2019; 43:12-17. [PMID: 30747024 DOI: 10.1080/03630269.2019.1567528] [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: 10/27/2022]
Abstract
Thalassemia is a hereditary disease with an autosomal recessive inheritance pattern resulting in reduced production of globin chains. Mutations in modifier genes can cause or affect thalassemia. Krüppel-like factor 1 (KLF1) is a modifier gene that was investigated in this study. Thirty-five Iranian β-thalassemia (β-thal) minor patients with hematological symptoms including Hb A2 3.0%, mean corpuscular volume (MCV) <75.0 fL, mean corpuscular hemoglobin (Hb) (MCH) <25.0 pg, and two β-thal intermedia (β-TI) patients in 50 subjects who carried no mutations on the HBB and HBA2 or HBA1 genes were investigated for all exons of the KLF1 gene by polymerase chain reaction (PCR) and sequencing methods. Of the 35 patients with a β-thal minor phenotype, one patient was heterozygous for the c.544T>C mutation in exon 2 of KLF1 and HBB: c.380T>G variant, Hb Dhonburi [also known as Hb Neapolis or codon 126 (T>G)]. The c.340T>C mutation was also found in exon 2 of the KLF1 gene with an allele frequency of 16.6% in the studied β-thal carriers. The two β-TI patients were homozygous for a new mutation c.942delA in exon 3 of KLF1. Mutations in modifier genes can cause or affect thalassemia. Therefore, exact investigation of globin genes and modifiers such as KLF1 is necessary in areas where globin gene disorders are most prevalent to understand the reason of clinical and hematological symptoms of thalassemia and facilitate newborn screening or prenatal diagnosis (PND) programs.
Collapse
Affiliation(s)
- Ahmad Tamaddoni
- a Non-Communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences , Babol , Iran
| | - Sahar Khabaz Astaneh
- b Department of Cellular and Molecular Biology , Islamic Azad University , Babol Branch , Babol , Iran
| | - Reza Tabaripour
- b Department of Cellular and Molecular Biology , Islamic Azad University , Babol Branch , Babol , Iran
| | - Haleh Akhavan-Niaki
- c Department of Genetics, Faculty of Medicine , Babol University of Medical Sciences , Babol , Iran
| |
Collapse
|
71
|
Chaweephisal P, Phusua A, Fanhchaksai K, Sirichotiyakul S, Charoenkwan P. Borderline hemoglobin A2 levels in northern Thai population: HBB genotypes and effects of coinherited alpha-thalassemia. Blood Cells Mol Dis 2019; 74:13-17. [DOI: 10.1016/j.bcmd.2018.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 01/13/2023]
|
72
|
Ducamp S, Fleming MD. The molecular genetics of sideroblastic anemia. Blood 2019; 133:59-69. [PMID: 30401706 PMCID: PMC6318428 DOI: 10.1182/blood-2018-08-815951] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/21/2018] [Indexed: 01/19/2023] Open
Abstract
The sideroblastic anemias (SAs) are a group of inherited and acquired bone marrow disorders defined by pathological iron accumulation in the mitochondria of erythroid precursors. Like most hematological diseases, the molecular genetic basis of the SAs has ridden the wave of technology advancement. Within the last 30 years, with the advent of positional cloning, the human genome project, solid-state genotyping technologies, and next-generation sequencing have evolved to the point where more than two-thirds of congenital SA cases, and an even greater proportion of cases of acquired clonal disease, can be attributed to mutations in a specific gene or genes. This review focuses on an analysis of the genetics of these diseases and how understanding these defects may contribute to the design and implementation of rational therapies.
Collapse
Affiliation(s)
- Sarah Ducamp
- Department of Pathology, Boston Children's Hospital, Boston, MA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA
| |
Collapse
|
73
|
Ma H, Davarifar A, Amengual JE. The Future of Combination Therapies for Peripheral T Cell Lymphoma (PTCL). Curr Hematol Malig Rep 2018; 13:13-24. [PMID: 29397528 DOI: 10.1007/s11899-018-0432-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Peripheral T cell lymphoma is a rare heterogeneous group of diseases which are characterized by poor outcomes to treatment and short overall survival. In the past decade, several new therapies targeting T cell biology have been approved in the relapsed setting. These new therapies, such as pralatrexate, romidepsin, belinostat, and brentuximab vedotin, have begun to make their way into practice. Despite these advances, outcomes have not changed dramatically. In recent years, efforts have been made to incorporate these new therapies into combination strategies to treat this challenging disease entity. Herein we will review some of the latest developments. RECENT FINDINGS With the new WHO classification, discrete entities of PTCL are now being identified by molecular and phenotypic markers. This new classification is critical to our ability to define disease entities which may respond to certain classes of targeted therapy. Some such mutations include genes controlling epigenetics (TET2, IDH2, DNMT3A, RHOA, CD28). As such, epigenetic therapies such as histone deacetylase (HDAC) inhibitors have become the platform to which other novel therapies or chemotherapy has been added. Early phase clinical studies have demonstrated that combination therapy with romidepsin plus other agents known to have activity in T cell lymphoma have enhanced clinical benefit for this group of diseases. In addition, the antibody drug conjugate, brentuximab vedotin has been shown to have potent activity in T cell lymphomas expressing CD30. This drug is being studied as well with other targeted therapies and chemotherapy in an effort to improve response rates and progression-free survival. Although T cell lymphomas remain a highly challenging group of diseases to treat, new efforts to leverage drugs that discretely target the biology that drives T cell lymphomagenesis in combination provide hope that improved outcomes may be realized in the near future.
Collapse
Affiliation(s)
- Helen Ma
- Center for Lymphoid Malignancies, Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, 51 West 51st Street, Suite 200, New York, NY, 10019, USA
| | - Ardy Davarifar
- Center for Lymphoid Malignancies, Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, 51 West 51st Street, Suite 200, New York, NY, 10019, USA
| | - Jennifer E Amengual
- Center for Lymphoid Malignancies, Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, 51 West 51st Street, Suite 200, New York, NY, 10019, USA.
| |
Collapse
|
74
|
Hariharan P, Colah R, Ghosh K, Nadkarni A. Differential role of Kruppel like factor 1 (KLF1) gene in red blood cell disorders. Genomics 2018; 111:1771-1776. [PMID: 30529538 DOI: 10.1016/j.ygeno.2018.11.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023]
Abstract
The master erythroid regulator KLF1,plays a pivotal role during erythroid lineage development by regulating the expression of many erythroid genes. Variations in the KLF1 gene are found to be associated with varied erythroid phenotypes. With the aim of determining the role of KLF1 gene variations in HbF induction and their genotype phenotype relationship, in this study, we screened 370 individuals with different hemoglobinopathy condition. Hematological analysis was carried out using automated blood cell counter and Variant II HPLC (Biorad). KLF1 gene mutations were screened using automated DNA sequencing. Expression analysis was carried out using q-RT PCR of KLF1, BCL11A and γ-globin after selective enrichment and culturing of CD 34 +ve cells into an erythroid lineage. Over all 14 KLF1 gene variations were identified, of which six variants were novel. The incidence of KLF1 gene mutations was found to be 8.1%. It was seen that KLF1 mutations contributed in borderline HbA2 levels as 7.6% of our borderline HbA2 cases showed presence of KLF1 variations. It also contributed in induction of HbF levels under stress erythropoietic conditions. Gene expression studies revealed inverse correlation of KLF1, BCL11A (reduced) with γ-globin gene expression (increased) in patients showing KLF1 gene mutations, thus indicating the role of KLF1 gene in regulating the γ-globin gene expression. The identification of genomic variants of the KLF1 may help in determining the functionally active domain of this protein and will facilitate in understanding the wide spectrum of phenotypes generated by these variants.
Collapse
Affiliation(s)
- Priya Hariharan
- National Institute of Immunohematology (ICMR), 13th Floor, New Multi-storeyed Building, K.E.M. Hospital Campus, Parel, Mumbai 400012, India
| | - Roshan Colah
- National Institute of Immunohematology (ICMR), 13th Floor, New Multi-storeyed Building, K.E.M. Hospital Campus, Parel, Mumbai 400012, India
| | - Kanjaksha Ghosh
- National Institute of Immunohematology (ICMR), 13th Floor, New Multi-storeyed Building, K.E.M. Hospital Campus, Parel, Mumbai 400012, India
| | - Anita Nadkarni
- National Institute of Immunohematology (ICMR), 13th Floor, New Multi-storeyed Building, K.E.M. Hospital Campus, Parel, Mumbai 400012, India.
| |
Collapse
|
75
|
Pu J, Zhang L, Wei X, Xu X. Clinical Genotyping by Next Generation Sequencing Reveals a Novel, De Novo β-Globin Gene Mutation Causing Hemolytic Anemia in a Chinese Individual. Hemoglobin 2018; 42:184-188. [DOI: 10.1080/03630269.2018.1496928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Jiajie Pu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Li Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xiaofeng Wei
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
76
|
Jiang F, Qu YX, Chen GL, Li J, Zhou JY, Zuo LD, Liao C, Li DZ. KFL1 Gene Variants in α-Thalassemia Individuals with Increased Fetal Hemoglobin in a Chinese Population. Hemoglobin 2018; 42:161-165. [PMID: 30205725 DOI: 10.1080/03630269.2018.1486325] [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] [Indexed: 10/28/2022]
Abstract
Krüppel-like factor 1 (KLF1) is a pleiotropic erythroid transcription factor that is a regulator of definitive erythropoiesis. The aim of this study was to detect KLF1 gene variants in α-thalassemia (α-thal) carriers with an increased Hb F level in a Chinese population, and determine the changes of hematological parameters as a result of interactions between KLF1 gene mutations and α-thal. Subjects with α-thal and Hb F levels of ≥1.0% were selected for further investigation. Direct sequencing was used to detect KLF1 gene mutations. Hematological parameters of subjects with α-thal and concomitant KLF1 gene mutations and those with α-thal alone were compared. The KLF1 gene variants were detected in 46 of 275 (16.7%) individuals with α-thal and Hb F levels of ≥1.0%. The detection rate of KLF1 gene mutations rose correspondingly when the Hb F level increased. For α0-thal carriers, significantly lower mean corpuscular volume (MCV) and mean corpuscular hemoglobin (Hb) (MCH) values were observed in KLF1 gene mutation-positive carriers than that in KLF1 gene mutation-free carriers; conversely, significantly higher Hb A2 and Hb F levels were observed in the former condition rather than in the latter condition. The results of this study indicate that KLF1 gene variants are common in Chinese subjects with α-thal and increased Hb F levels, and KLF1 gene mutations decreased the red blood cell (RBC) indices in α-thal carriers as that in normal adults.
Collapse
Affiliation(s)
- Fan Jiang
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Yan-Xia Qu
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Gui-Lan Chen
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Jian Li
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Jian-Ying Zhou
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Lian-Dong Zuo
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Can Liao
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Dong-Zhi Li
- a Guangzhou Women and Children's Medical Center , Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| |
Collapse
|
77
|
Rani N, Jamwal M, Kaur J, Sharma P, Malhotra P, Maitra A, Singh R, Das R. Homozygous KLF1 mutation c.901C>T (p.Arg301Cys) resulting in mild thalassemia intermedia in an Indian: A next-generation sequencing diagnosis. Blood Cells Mol Dis 2018; 72:19-21. [PMID: 29980343 DOI: 10.1016/j.bcmd.2018.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/18/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Neetu Rani
- Department cum National Centre for Human Genomics studies &Research, Panjab University, Sector 14, Chandigarh 160014, India
| | - Manu Jamwal
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Jasbir Kaur
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Prashant Sharma
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Pankaj Malhotra
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Arindam Maitra
- National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Ranvir Singh
- Department cum National Centre for Human Genomics studies &Research, Panjab University, Sector 14, Chandigarh 160014, India
| | - Reena Das
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
78
|
Tang HS, Wang DG, Huang LY, Li DZ. δ-Thalassemia with Complete Absence of Hb A 2 in a Chinese Family. Hemoglobin 2018; 42:135-137. [PMID: 29737888 DOI: 10.1080/03630269.2018.1463916] [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] [Indexed: 10/17/2022]
Abstract
A Chinese family with δ-thalassemia (δ-thal) was found, in which the daughter is homozygous for δ-thal (HBD: c.-127T>C) with complete deficiency of Hb A2 and the mother is a heterozygote with low level of Hb A2. The father, however, is a heterozygote with a normal Hb A2 value due to coinheritance of a β-thalassemia (β-thal). Although no abnormal clinical or hematological findings were noted in the individuals with δ-thal, one should keep in mind that β-thal can be missed during routine preliminary screening when β-thal and δ-thal coexist in a subject.
Collapse
Affiliation(s)
- Hai-Shen Tang
- a Prenatal Diagnosis Unit , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , People's Republic of China
| | - De-Gang Wang
- a Prenatal Diagnosis Unit , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , People's Republic of China
| | - Lv-Yin Huang
- b Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| | - Dong-Zhi Li
- b Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou , Guangdong Province , People's Republic of China
| |
Collapse
|
79
|
Gnanapragasam MN, Crispino JD, Ali AM, Weinberg R, Hoffman R, Raza A, Bieker JJ. Survey and evaluation of mutations in the human KLF1 transcription unit. Sci Rep 2018; 8:6587. [PMID: 29700354 PMCID: PMC5920080 DOI: 10.1038/s41598-018-24962-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023] Open
Abstract
Erythroid Krüppel-like Factor (EKLF/KLF1) is an erythroid-enriched transcription factor that plays a global role in all aspects of erythropoiesis, including cell cycle control and differentiation. We queried whether its mutation might play a role in red cell malignancies by genomic sequencing of the KLF1 transcription unit in cell lines, erythroid neoplasms, dysplastic disorders, and leukemia. In addition, we queried published databases from a number of varied sources. In all cases we only found changes in commonly notated SNPs. Our results suggest that if there are mutations in KLF1 associated with erythroid malignancies, they are exceedingly rare.
Collapse
Affiliation(s)
- Merlin Nithya Gnanapragasam
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - John D Crispino
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Abdullah M Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Rona Weinberg
- Cellular Therapy Laboratory, New York Blood Center, New York, NY, 10065, USA
| | - Ronald Hoffman
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - Azra Raza
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Black Familly Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
| |
Collapse
|
80
|
Abstract
Thalassemia is a disorder of hemoglobin characterized by reduced or absent production of one of the globin chains in human red blood cells with relative excess of the other. Impaired synthesis of β-globin results in β-thalassemia, whereas defective synthesis of α-globin leads to α-thalassemia. Despite being a monogenic disorder, thalassemia exhibits remarkable clinical heterogeneity that is directly related to the intracellular imbalance between α- and β-like globin chains. Novel insights into the genetic modifiers have contributed to the understanding of the correlation between genotype and phenotype and are being explored as therapeutic pathways to cure this life-limiting disease.
Collapse
Affiliation(s)
- Sachith Mettananda
- Molecular Hematology Unit, Medical Research Council (MRC), Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama 11010, Sri Lanka
| | - Douglas R Higgs
- Molecular Hematology Unit, Medical Research Council (MRC), Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; National Institute for Health Research, Oxford Biomedical Research Centre, Blood Theme, Oxford University Hospitals, Headington, Oxford OX3 9DU, UK.
| |
Collapse
|
81
|
Hamid M, Ershadi Oskouei S, Shariati G, Babaei E, Galehdari H, Saberi A, Sedaghat A. Mutation Screening of the Krüppel-like Factor 1 Gene in Individuals With Increased Fetal Hemoglobin Referred for Hemoglobinopathy Investigation in South of Iran. J Pediatr Hematol Oncol 2018; 40:192-195. [PMID: 29420372 DOI: 10.1097/mph.0000000000001093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Any mutation in the Krüppel-like factor 1 (KLF1) gene may interfere with its proper related function in the erythropoiesis process and lead to alterations in proper activation of its downstream protein through globin switching, which results in an increase in fetal hemoglobin (HbF). This study aimed to investigate whether KLF1 mutation can associate with high level of HbF in individuals with increased fetal hemoglobin referred for screening of hemoglobinopathies in south of Iran. MATERIALS AND METHODS The human KLF1 gene was amplified via the polymerase chain reaction procedure, and sequencing was used to determine any mutation in these patients. Moreover, XmnI polymorphisms in the position of -158 of γ-globin gene promoter were analyzed in all patients by polymerase chain reaction restriction fragment length polymorphism. RESULT Analysis of sequencing revealed a missense mutation in the KLF1 gene, p.Ser102Pro (c.304T>C), which was detectable in 10 of 23 cases with elevated HbF level. This mutation was only detected in individuals who had a HbF level between 3.1% and 25.6%. Statistical analysis showed that the frequency of C allele is significantly correlated with a high level of HbF (P<0.05). The allele frequency of positive result of XmnI polymorphism in individuals with increased HbF level was also significant, which showed an association with increased HbF level (P<0.05). CONCLUSIONS To the best of our knowledge, this is the first report of p.Ser102Pro (c.304T>C) in the KLF1 gene in β-thalassemia patients with increased level of fetal hemoglobin. According to statistical results of p.Ser102Pro mutation and XmnI polymorphism, it has been strongly suggested that both polymorphisms have an association with increased HbF samples. These nucleotide changes alone may not be the only elements raising the level of HbF, and other regulatory and modifying factors also play a role in HbF production.
Collapse
Affiliation(s)
- Mohammad Hamid
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran
| | | | - Gholamreza Shariati
- Department of Medical Genetics, Faculty of Medicine.,Narges Medical Genetics & PND Laboratory, Kianpars, Ahvaz, Iran
| | - Esmaeil Babaei
- Department of Biology, School of Natural Science, University of Tabriz, Tabriz
| | | | | | - Alireza Sedaghat
- Health Research Institute, Diabetes Research Center, Ahvaz Jundishapur University of Medical Sciences.,Narges Medical Genetics & PND Laboratory, Kianpars, Ahvaz, Iran
| |
Collapse
|
82
|
Li Y, Liu D, Zhang X, Li Z, Ye Y, Liu Q, Shen J, Chen Z, Huang H, Liang Y, Han X, Liu J, An X, Mohandas N, Xu X. miR-326 regulates HbF synthesis by targeting EKLF in human erythroid cells. Exp Hematol 2018; 63:33-40.e2. [PMID: 29601850 DOI: 10.1016/j.exphem.2018.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/27/2022]
Abstract
Haploinsufficiency of erythroid Krüppel-like factor (EKLF/KLF1) has been shown recently to ameliorate the clinical severity of β-thalassemia by increased expression levels of fetal hemoglobin (HbF). The underlying mechanisms for role of EKLF in regulating HbF are of great interest but remain incompletely understood. In this study, we used a combination of in silico, in vitro, and in vivo approaches to identify microRNAs (miRs) involved in EKLF regulation and to validate the role of miR-326 in HbF modification. We found that miR-326 suppresses EKLF expression directly by targeting its 3' untranslated region. miR-326 overexpression in K562 cells or CD34+ hematopoietic progenitor cells resulted in reduced EKLF protein levels and was associated with elevated expression of γ-globin, whereas inhibition of physiological miR-326 levels increased EKLF and thus reduced γ-globin expression. Moreover, miR-326 expression is positively correlated with HbF levels in β-thalassemia patients. Our results suggest that miR-326 plays a key role in regulating EKLF expression and in modifying the HbF level, which may provide a new strategy for activating HbF in individuals with β-thalassemia or sickle cell disease.
Collapse
Affiliation(s)
- Yihong Li
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Dun Liu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinhua Zhang
- Department of Hematology, 303rd Hospital of the People's Liberation Army, Nanning, Guangxi, China
| | - Zhiming Li
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuhua Ye
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Shen
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi Chen
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huajie Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunhao Liang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xu Han
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jing Liu
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA; College of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, USA
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
83
|
Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol 2018; 15:273-291. [PMID: 29508857 DOI: 10.1038/nrclinonc.2018.28] [Citation(s) in RCA: 705] [Impact Index Per Article: 117.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The PI3K-AKT-mTOR pathway is one of the most frequently dysregulated pathways in cancer and, consequently, more than 40 compounds that target key components of this signalling network have been tested in clinical trials involving patients with a range of different cancers. The clinical development of many of these agents, however, has not advanced to late-phase randomized trials, and the antitumour activity of those that have been evaluated in comparative prospective studies has typically been limited, or toxicities were found to be prohibitive. Nevertheless, the mTOR inhibitors temsirolimus and everolimus and the PI3K inhibitors idelalisib and copanlisib have been approved by the FDA for clinical use in the treatment of a number of different cancers. Novel compounds with greater potency and selectivity, as well as improved therapeutic indices owing to reduced risks of toxicity, are clearly required. In addition, biomarkers that are predictive of a response, such as PIK3CA mutations for inhibitors of the PI3K catalytic subunit α isoform, must be identified and analytically and clinically validated. Finally, considering that oncogenic activation of the PI3K-AKT-mTOR pathway often occurs alongside pro-tumorigenic aberrations in other signalling networks, rational combinations are also needed to optimize the effectiveness of treatment. Herein, we review the current experience with anticancer therapies that target the PI3K-AKT-mTOR pathway.
Collapse
Affiliation(s)
- Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
84
|
Huang LY, Li J, Zhang Y, Li DZ. A KLF1
gene mutation causes β-thalassemia minor in a Chinese family. Int J Lab Hematol 2018; 40:e35-e37. [DOI: 10.1111/ijlh.12788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/16/2018] [Indexed: 11/29/2022]
Affiliation(s)
- L.-Y. Huang
- Guangzhou Women and Children Medical Center affiliated to Guangzhou Medical University; Guangzhou Guangdong China
| | - J. Li
- Guangzhou Women and Children Medical Center affiliated to Guangzhou Medical University; Guangzhou Guangdong China
| | | | - D.-Z. Li
- Guangzhou Women and Children Medical Center affiliated to Guangzhou Medical University; Guangzhou Guangdong China
| |
Collapse
|
85
|
Asadov C, Alimirzoeva Z, Mammadova T, Aliyeva G, Gafarova S, Mammadov J. β-Thalassemia intermedia: a comprehensive overview and novel approaches. Int J Hematol 2018; 108:5-21. [PMID: 29380178 DOI: 10.1007/s12185-018-2411-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/19/2018] [Accepted: 01/24/2018] [Indexed: 01/19/2023]
Abstract
β-Thalassemia intermedia is a clinical condition of intermediate gravity between β-thalassemia minor, the asymptomatic carrier, and β-thalassemia major, the transfusion-dependent severe anemia. It is characterized by a significant clinical polymorphism, which is attributable to its genetic heterogeneity. Ineffective erythropoiesis, chronic anemia, and iron overload contribute to the clinical complications of thalassemia intermedia through stepwise pathophysiological mechanisms. These complications, including splenomegaly, extramedullary erythropoiesis, iron accumulation, leg ulcers, thrombophilia, and bone abnormalities can be managed via fetal hemoglobin induction, occasional transfusions, chelation, and in some cases, stem cell transplantation. Given its clinical diversity, thalassemia intermedia patients require tailored approaches to therapy. Here we present an overview and novel approaches to the genetic basis, pathophysiological mechanisms, clinical complications, and optimal management of thalassemia intermedia.
Collapse
Affiliation(s)
- Chingiz Asadov
- Institute of Hematology and Transfusiology, M. Gashgai Str. 87, AZ1007, Baku, Azerbaijan.
| | - Zohra Alimirzoeva
- Institute of Hematology and Transfusiology, M. Gashgai Str. 87, AZ1007, Baku, Azerbaijan
| | - Tahira Mammadova
- Institute of Hematology and Transfusiology, M. Gashgai Str. 87, AZ1007, Baku, Azerbaijan
| | - Gunay Aliyeva
- Institute of Hematology and Transfusiology, M. Gashgai Str. 87, AZ1007, Baku, Azerbaijan
| | - Shahla Gafarova
- Institute of Hematology and Transfusiology, M. Gashgai Str. 87, AZ1007, Baku, Azerbaijan
| | - Jeyhun Mammadov
- Thalassemia Centre, Fataly Khan Khoysky Str. 128, AZ1072, Baku, Azerbaijan
| |
Collapse
|
86
|
Janiga J, Kentley J, Nabhan C, Abdulla F. Current systemic therapeutic options for advanced mycosis fungoides and Sézary syndrome. Leuk Lymphoma 2018; 59:562-577. [PMID: 29308723 DOI: 10.1080/10428194.2017.1347650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are the most common cutaneous T-cell lymphomas (CTCLs). Both lack curative options, and advanced-stage carries a poor prognosis. Whilst there are a number of treatments available, achieving and maintaining a durable remission remains challenging. We review current systemic treatment options as monotherapy for advanced-stage MF (IIB-IV), appraising their mechanism of action, analyzing their efficacy, and describing toxicities. Individually, reported overall response rates (ORR) vary widely in the literature and duration of responses are typically short, ranging from 7.5 to 22.4 months. Combined therapy is frequently used in an effort to boost responses, although prospective studies comparing combinations to single agent therapies are rarely conducted. While recent translational research has led to increased understanding of the immunopathogenesis of MF and SS and the development of new treatments, current standard of care therapies are not curative and have low ORR for advanced-stage disease.
Collapse
Affiliation(s)
- Jenna Janiga
- a Stritch School of Medicine , Loyola University , Chicago , IL , USA
| | - Jonathan Kentley
- b Department of Dermatology , Royal London Hospital, Barts Health NHS Trust , London , UK
| | - Chadi Nabhan
- c Cardinal Health Specialty Solutions , Waukegan , IL , USA
| | - Farah Abdulla
- d Department of Medicine, Section of Dermatology , University of Chicago Medicine and Biological Sciences , Chicago , IL , USA
| |
Collapse
|
87
|
|
88
|
Borgio JF, AbdulAzeez S, Al-Muslami AM, Naserullah ZA, Al-Jarrash S, Al-Suliman AM, Al-Madan MS, Al-Ali AK. KLF1 gene and borderline hemoglobin A 2 in Saudi population. Arch Med Sci 2018; 14:230-236. [PMID: 29379553 PMCID: PMC5778434 DOI: 10.5114/aoms.2018.72245] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/29/2017] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Elevated HbA2 (hemoglobin A2) level is considered the most reliable hematological parameter for the detection of β-thalassemia carriers. However, some carriers are difficult to recognize because the level of HbA2 is not in the distinctive carrier range, i.e. 4.0-6.0%; instead, some carriers have HbA2 levels between normal and carrier levels, i.e. borderline HbA2 (HbA2 = 3.1-3.9%). Studies have shown that variations in the erythroid Krüppel-like factor (KLF1) gene lead to borderline HbA2 in β-thalassemia carriers from various populations. The incidence of borderline HbA2 in Saudis is high. MATERIAL AND METHODS To confirm the influence of variations in KLF1, HBA1, HBA2 and HBB genes for the reduction of the level of HbA2 in Saudi β-thalassemia carriers, we performed a direct sequence analysis of KLF1, HBA1, HBA2 and HBB genes from 212 healthy Saudis (88 subjects: HbA2 < 3; 72 subjects: HbA2 = 3.1 to 3.9; 52 subjects HbA2 > 4.3). RESULTS The presence of the borderline HbA2 level is not specific to any type of β-thalassemia variation or β+-thalassemia variations in Saudis. Two exonic (c.304T>C and c.544T>C) and two 3' untranslated region (3'UTR) (c.*296G>A and c.*277C>G) variations have been identified in the KLF1 gene for the first time from an Arab population. None of these four variations in KLF1 genes are significantly associated with the Saudis with borderline HbA2. α Globin genotype, -α23.7/α1α2, is found to be the most frequent (55.55%) among healthy Saudis with borderline HbA2 compared with the other groups (HbA2 < 3 = 20.45%; HbA2 > 4.3 = 13.51%). CONCLUSIONS Further studies are necessary to determine the influence of other factors on the presence of borderline HbA2 in 41.67% of Saudis.
Collapse
Affiliation(s)
- J. Francis Borgio
- Department of Genetic Research, Institute for Research and Medical Consultation (IRMC), University of Dammam, Dammam, Saudi Arabia
| | - Sayed AbdulAzeez
- Department of Genetic Research, Institute for Research and Medical Consultation (IRMC), University of Dammam, Dammam, Saudi Arabia
| | - Ahmed M. Al-Muslami
- Department of Genetic Research, Institute for Research and Medical Consultation (IRMC), University of Dammam, Dammam, Saudi Arabia
| | | | | | - Ahmed M. Al-Suliman
- Al-Omran Scientific Chair for Hematological Diseases Prevalent in the Al-Ahssa Area, King Faisal University, Al-Ahssa, Saudi Arabia
| | - Mohammed S. Al-Madan
- Department of Pediatrics, King Fahd Hospital of the University, Al-Khobar, Saudi Arabia
| | - Amein K. Al-Ali
- Department of Biochemistry, University of Dammam, Dammam, Saudi Arabia
| |
Collapse
|
89
|
Chapin J, Giardina PJ. Thalassemia Syndromes. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00040-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
90
|
Vinjamur DS, Bauer DE, Orkin SH. Recent progress in understanding and manipulating haemoglobin switching for the haemoglobinopathies. Br J Haematol 2017; 180:630-643. [PMID: 29193029 DOI: 10.1111/bjh.15038] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The major β-haemoglobinopathies, sickle cell disease and β-thalassaemia, represent the most common monogenic disorders worldwide and a steadily increasing global disease burden. Allogeneic haematopoietic stem cell transplantation, the only curative therapy, is only applied to a small minority of patients. Common clinical management strategies act mainly downstream of the root causes of disease. The observation that elevated fetal haemoglobin expression ameliorates these disorders has motivated longstanding investigations into the mechanisms of haemoglobin switching. Landmark studies over the last decade have led to the identification of two potent transcriptional repressors of γ-globin, BCL11A and ZBTB7A. These regulators act with additional trans-acting epigenetic repressive complexes, lineage-defining factors and developmental programs to silence fetal haemoglobin by working on cis-acting sequences at the globin gene loci. Rapidly advancing genetic technology is enabling researchers to probe deeply the interplay between the molecular players required for γ-globin (HBG1/HBG2) silencing. Gene therapies may enable permanent cures with autologous modified haematopoietic stem cells that generate persistent fetal haemoglobin expression. Ultimately rational small molecule pharmacotherapies to reactivate HbF could extend benefits widely to patients.
Collapse
Affiliation(s)
- Divya S Vinjamur
- Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Stuart H Orkin
- Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
91
|
Genetic variation of Krüppel-like factor 1 (KLF1) and fetal hemoglobin (HbF) levels in β 0-thalassemia/HbE disease. Int J Hematol 2017; 107:297-310. [PMID: 29067594 DOI: 10.1007/s12185-017-2357-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022]
Abstract
Heterogeneity of HbF levels in β0-thalassemia/HbE disease has been reported to be associated with variations in clinical manifestations of the disease, and several genetic-modifying factors beyond the β-globin gene cluster have been identified as HbF regulators. Down-regulation or heterozygous mutations of Krüppel-like factor 1 (KLF1) is associated with elevated HbF levels in non-thalassemia subjects. This study confirms that experimental down-regulation of KLF1 in β0-thalassemia/HbE-derived erythroblasts significantly increases HbF production (up to 52.3 ± 2.4%), albeit with slightly delayed erythroid terminal differentiation. KLF1 exome sequencing of 130 Thai β0-thalassemia/HbE patients without co-inheritance of α-thalassemia found six patients with KLF1 heterozygous mutations including rs2072596 (p.F182L; n = 5) and rs745347362 (p.P284L; n = 1) missense mutations. However, while these patients had high HbF levels (38.1 ± 7.5%), they were all associated with a severe clinical phenotype. These results suggest that while reduction of KLF1 expression in β0-thalassemia/HbE erythroblasts can increase HbF levels, it is not sufficient to alleviate the clinical phenotype.
Collapse
|
92
|
He Y, Luo J, Chen Y, Zhou X, Yu S, Jin L, Xiao X, Jia S, Liu Q. ARHGAP18 is a novel gene under positive natural selection that influences HbF levels in β-thalassaemia. Mol Genet Genomics 2017; 293:207-216. [DOI: 10.1007/s00438-017-1377-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
|
93
|
Gillinder KR, Ilsley MD, Nébor D, Sachidanandam R, Lajoie M, Magor GW, Tallack MR, Bailey T, Landsberg MJ, Mackay JP, Parker MW, Miles LA, Graber JH, Peters LL, Bieker JJ, Perkins AC. Promiscuous DNA-binding of a mutant zinc finger protein corrupts the transcriptome and diminishes cell viability. Nucleic Acids Res 2017; 45:1130-1143. [PMID: 28180284 PMCID: PMC5388391 DOI: 10.1093/nar/gkw1014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/13/2016] [Accepted: 11/02/2016] [Indexed: 12/27/2022] Open
Abstract
The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.
Collapse
Affiliation(s)
- Kevin R Gillinder
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Melissa D Ilsley
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | | | - Ravi Sachidanandam
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Mathieu Lajoie
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Graham W Magor
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Michael R Tallack
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Timothy Bailey
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, USA
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, NSW, Australia
| | - Michael W Parker
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia.,ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Luke A Miles
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, USA
| | - Andrew C Perkins
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| |
Collapse
|
94
|
Planutis A, Xue L, Trainor CD, Dangeti M, Gillinder K, Siatecka M, Nebor D, Peters LL, Perkins AC, Bieker JJ. Neomorphic effects of the neonatal anemia (Nan-Eklf) mutation contribute to deficits throughout development. Development 2017; 144:430-440. [PMID: 28143845 DOI: 10.1242/dev.145656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/18/2016] [Indexed: 12/20/2022]
Abstract
Transcription factor control of cell-specific downstream targets can be significantly altered when the controlling factor is mutated. We show that the semi-dominant neonatal anemia (Nan) mutation in the EKLF/KLF1 transcription factor leads to ectopic expression of proteins that are not normally expressed in the red blood cell, leading to systemic effects that exacerbate the intrinsic anemia in the adult and alter correct development in the early embryo. Even when expressed as a heterozygote, the Nan-EKLF protein accomplishes this by direct binding and aberrant activation of genes encoding secreted factors that exert a negative effect on erythropoiesis and iron use. Our data form the basis for a novel mechanism of physiological deficiency that is relevant to human dyserythropoietic anemia and likely other disease states.
Collapse
Affiliation(s)
- Antanas Planutis
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Li Xue
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Cecelia D Trainor
- Laboratory of Molecular Biology, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Mohan Dangeti
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Kevin Gillinder
- Mater Research Institute, University of Queensland, Woolloongabba QLD 4102, Queensland, Australia
| | - Miroslawa Siatecka
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.,Department of Genetics, University of Adam Mickiewicz, Poznan 61-614, Poland
| | | | | | - Andrew C Perkins
- Mater Research Institute, University of Queensland, Woolloongabba QLD 4102, Queensland, Australia.,Princess Alexandra Hospital, Brisbane QLD 4102, Queensland, Australia
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA .,Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.,Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.,Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
95
|
Shang X, Peng Z, Ye Y, Asan, Zhang X, Chen Y, Zhu B, Cai W, Chen S, Cai R, Guo X, Zhang C, Zhou Y, Huang S, Liu Y, Chen B, Yan S, Chen Y, Ding H, Yin X, Wu L, He J, Huang D, He S, Yan T, Fan X, Zhou Y, Wei X, Zhao S, Cai D, Guo F, Zhang Q, Li Y, Zhang X, Lu H, Huang H, Guo J, Zhu F, Yuan Y, Zhang L, Liu N, Li Z, Jiang H, Zhang Q, Zhang Y, Juhari WKW, Hanafi S, Zhou W, Xiong F, Yang H, Wang J, Zilfalil BA, Qi M, Yang Y, Yin Y, Mao M, Xu X. Rapid Targeted Next-Generation Sequencing Platform for Molecular Screening and Clinical Genotyping in Subjects with Hemoglobinopathies. EBioMedicine 2017; 23:150-159. [PMID: 28865746 PMCID: PMC5605365 DOI: 10.1016/j.ebiom.2017.08.015] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/15/2017] [Accepted: 08/15/2017] [Indexed: 12/17/2022] Open
Abstract
Hemoglobinopathies are among the most common autosomal-recessive disorders worldwide. A comprehensive next-generation sequencing (NGS) test would greatly facilitate screening and diagnosis of these disorders. An NGS panel targeting the coding regions of hemoglobin genes and four modifier genes was designed. We validated the assay by using 2522 subjects affected with hemoglobinopathies and applied it to carrier testing in a cohort of 10,111 couples who were also screened through traditional methods. In the clinical genotyping analysis of 1182 β-thalassemia subjects, we identified a group of additional variants that can be used for accurate diagnosis. In the molecular screening analysis of the 10,111 couples, we detected 4180 individuals in total who carried 4840 mutant alleles, and identified 186 couples at risk of having affected offspring. 12.1% of the pathogenic or likely pathogenic variants identified by our NGS assay, which were undetectable by traditional methods. Compared with the traditional methods, our assay identified an additional at-risk 35 couples. We describe a comprehensive NGS-based test that offers advantages over the traditional screening/molecular testing methods. To our knowledge, this is among the first large-scale population study to systematically evaluate the application of an NGS technique in carrier screening and molecular diagnosis of hemoglobinopathies.
Collapse
Affiliation(s)
- Xuan Shang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Zhiyu Peng
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China
| | - Yuhua Ye
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Asan
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Xinhua Zhang
- Department of Hematology, 303rd Hospital of the People's Liberation Army, Nanning, Guangxi, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Baosheng Zhu
- Genetic Diagnosis Center, First People's Hospital of Yunnan Province, Medical School of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wangwei Cai
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan, China
| | - Shaoke Chen
- Department of Genetic and Metabolic Laboratory, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, Guangxi, China
| | - Ren Cai
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Xiaoling Guo
- Maternity and Child Health Care Hospital of Foshan City, Foshan, Guangdong, China
| | - Chonglin Zhang
- Guilin Women and Children health care hospital, Guilin, Guangxi, China
| | - Yuqiu Zhou
- Department of Clinical Laboratory, Zhuhai Municipal Maternal and Child Healthcare Hospital, Zhuhai Institute of Medical Genetics, Zhuhai, Guangdong, China
| | - Shuodan Huang
- Maternal and Child Health Hospital in Meizhou, Meizhou, Guangdong, China
| | - Yanhui Liu
- Department of Prenatal Diagnosis Center, Dong Guan Maternal and Child Health Hospital, Dongguan, Guangdong, China
| | - Biyan Chen
- Baise Women and Children Care Hospital, Baise, Guangxi, China
| | - Shanhuo Yan
- Genetic Laboratory, Qinzhou Maternaland Child Health Hospital, Qingzhou, Guangxi, China
| | - Yajun Chen
- Women and Children's Health Hospital of Shaoguan, Shaoguan, Guangdong, China
| | - Hongmei Ding
- Department of Gynecology and Obstetrics, The People's Hospital of Yunfu City, Yunfu, Guangdong, China
| | - Xiaolin Yin
- Department of Hematology, 303rd Hospital of the People's Liberation Army, Nanning, Guangxi, China
| | - Liusong Wu
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Jing He
- Genetic Diagnosis Center, First People's Hospital of Yunnan Province, Medical School of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Dongai Huang
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan, China
| | - Sheng He
- Department of Genetic and Metabolic Laboratory, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, Guangxi, China
| | - Tizhen Yan
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Xin Fan
- Department of Genetic and Metabolic Laboratory, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, Guangxi, China
| | - Yuehong Zhou
- Pingguo Women and Children Care Hospital, Baise, Guangxi, China
| | - Xiaofeng Wei
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Sumin Zhao
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Decheng Cai
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Fengyu Guo
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Qianqian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Yun Li
- BGI Clinical Laboratories-Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Xuelian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Haorong Lu
- BGI Clinical Laboratories-Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Huajie Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Junfu Guo
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Fei Zhu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Yuan Yuan
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Li Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Na Liu
- BGI Clinical Laboratories-Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Zhiming Li
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Hui Jiang
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China
| | - Qiang Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Yijia Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | | | - Sarifah Hanafi
- Department of Paediatric, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Wanjun Zhou
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Fu Xiong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China
| | - Huanming Yang
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China; James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, China
| | - Jian Wang
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China; James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, China
| | - Bin Alwi Zilfalil
- Department of Paediatric, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Ming Qi
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Center for Genetic & Genomic Medicine, Zhejiang University Medical School 1st Affiliated Hospital, James Watson Institute of Genome Sciences, Hangzhou, Zhejiang, China
| | - Yaping Yang
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, USA
| | - Ye Yin
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China
| | - Mao Mao
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong, China.
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Biological Chip, Guangzhou, Guangdong, China.
| |
Collapse
|
96
|
Yu S, Chen Y, Lai K, Dewan RK, He Y. A Novel Variant with Positive Natural Selection Influenced Hb A 2 Levels in Chinese Individuals with β-Thalassemia. Hemoglobin 2017; 41:193-197. [PMID: 28747083 DOI: 10.1080/03630269.2017.1358177] [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: 10/19/2022]
Abstract
β-Thalassemia (β-thal) is the most common inherited hemolytic anemia worldwide. Elevated Hb A2 is a mark of β-thal carriers. The aim of this study was to identify the pathogenic variants associated with the Hb A2 levels. One thousand and thirty β-thal carriers were recruited for this study. Using positive natural expression quantitative trait loci (eQTL) analysis, a significant variant was selected. Genotyping for the rs231841 polymorphism was performed by the Sequenom MassARRAY IPLEX platform. All genetic association analyses were performed with the PLINK program. The linear regression analysis showed that rs231841 in the intron region of the potassium voltage-gated channel subfamily Q member 1 (KCNQ1) gene on chromosome 11p15 was significantly associated with Hb A2 levels. The presence of the C allele was associated with elevated Hb A2 levels. Our results suggest that rs231841 on the KCNQ1 gene with positive natural selection is related to Hb A2 levels in Chinese β-thal carriers, and KCNQ1 is probably associated with the expression of the β-like globin gene cluster.
Collapse
Affiliation(s)
- Shanjuan Yu
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , Guangxi Province , People's Republic of China
| | - Yang Chen
- b School of Public Health, Guangxi Medical University , Nanning , Guangxi Province , People's Republic of China
| | - Ketong Lai
- c Guangxi Key Laboratory of Thalassemia Research , The First Affiliated Hospital of Guangxi Medical University , Nanning , Guangxi Province , People's Republic of China
| | - Roma Kajal Dewan
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , Guangxi Province , People's Republic of China
| | - Yunyan He
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , Guangxi Province , People's Republic of China
| |
Collapse
|
97
|
Chen D, Zuo Y, Zhang X, Ye Y, Bao X, Huang H, Tepakhan W, Wang L, Ju J, Chen G, Zheng M, Liu D, Huang S, Zong L, Li C, Chen Y, Zheng C, Shi L, Zhao Q, Wu Q, Fucharoen S, Zhao C, Xu X. A Genetic Variant Ameliorates β-Thalassemia Severity by Epigenetic-Mediated Elevation of Human Fetal Hemoglobin Expression. Am J Hum Genet 2017; 101:130-138. [PMID: 28669403 DOI: 10.1016/j.ajhg.2017.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
A delayed fetal-to-adult hemoglobin (Hb) switch ameliorates the severity of β-thalassemia and sickle cell disease. The molecular mechanism underlying the epigenetic dysregulation of the switch is unclear. To explore the potential cis-variants responsible for the Hb switching, we systematically analyzed an 80-kb region spanning the β-globin cluster using capture-based next-generation sequencing of 1142 Chinese β-thalassemia persons and identified 31 fetal hemoglobin (HbF)-associated haplotypes of the selected 28 tag regulatory single-nucleotide polymorphisms (rSNPs) in seven linkage disequilibrium (LD) blocks. A Ly1 antibody reactive (LYAR)-binding motif disruptive rSNP rs368698783 (G/A) from LD block 5 in the proximal promoter of hemoglobin subunit gamma 1 (HBG1) was found to be a significant predictor for β-thalassemia clinical severity by epigenetic-mediated variant-dependent HbF elevation. We found this rSNP accounted for 41.6% of β-hemoglobinopathy individuals as an ameliorating factor in a total of 2,738 individuals from southern China and Thailand. We uncovered that the minor allele of the rSNP triggers the attenuation of LYAR and two repressive epigenetic regulators DNA methyltransferase 3 alpha (DNMT3A) and protein arginine methyltransferase 5 (PRMT5) from the HBG promoters, mediating allele-biased γ-globin elevation by facilitating demethylation of HBG core promoter CpG sites in erythroid progenitor cells from β-thalassemia persons. The present study demonstrates that this common rSNP in the proximal Aγ-promoter is a major genetic modifier capable of ameliorating the severity of thalassemia major through the epigenetic-mediated regulation of the delayed fetal-to-adult Hb switch and provides potential targets for the treatment of β-hemoglobinopathy.
Collapse
|
98
|
Vasseur C, Domingues-Hamdi E, Ledudal K, Le Corvoisier P, Barau C, Ghaleh B, Rialland A, Pissard S, Galactéros F, Baudin-Creuza V. Red blood cells free α-haemoglobin pool: a biomarker to monitor the β-thalassemia intermedia variability. The ALPHAPOOL study. Br J Haematol 2017. [DOI: 10.1111/bjh.14800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Corinne Vasseur
- Institut National de la Santé et de la Recherche Médicale (Inserm)-U955; équipe 2 : Transfusion et Maladies du Globule Rouge; Institut Mondor de Recherche Biomédicale (IMRB); Université de Paris Est Créteil (UPEC); Créteil France
- Laboratory of Excellence GR-Ex; Paris France
| | - Elisa Domingues-Hamdi
- Institut National de la Santé et de la Recherche Médicale (Inserm)-U955; équipe 2 : Transfusion et Maladies du Globule Rouge; Institut Mondor de Recherche Biomédicale (IMRB); Université de Paris Est Créteil (UPEC); Créteil France
- Laboratory of Excellence GR-Ex; Paris France
| | - Katia Ledudal
- Inserm; Centre d'Investigation Clinique 1430; Hôpital Universitaire Henri Mondor Assistance Publique Hôpitaux de Paris (AP-HP); Créteil France
| | - Philippe Le Corvoisier
- Inserm; Centre d'Investigation Clinique 1430; Hôpital Universitaire Henri Mondor Assistance Publique Hôpitaux de Paris (AP-HP); Créteil France
| | - Caroline Barau
- Plateforme de Ressources Biologiques; Hôpital Universitaire Henri Mondor (AP-HP); Créteil France
| | - Bijan Ghaleh
- Plateforme de Ressources Biologiques; Hôpital Universitaire Henri Mondor (AP-HP); Créteil France
| | - Amandine Rialland
- Unité de Recherche Clinique; Hôpital Universitaire Henri Mondor (AP-HP); Créteil France
| | - Serge Pissard
- Institut National de la Santé et de la Recherche Médicale (Inserm)-U955; équipe 2 : Transfusion et Maladies du Globule Rouge; Institut Mondor de Recherche Biomédicale (IMRB); Université de Paris Est Créteil (UPEC); Créteil France
- Laboratory of Excellence GR-Ex; Paris France
- Laboratoire de Génétique; Hôpital Universitaire Henri Mondor (AP-HP); Créteil France
| | - Frédéric Galactéros
- Institut National de la Santé et de la Recherche Médicale (Inserm)-U955; équipe 2 : Transfusion et Maladies du Globule Rouge; Institut Mondor de Recherche Biomédicale (IMRB); Université de Paris Est Créteil (UPEC); Créteil France
- Laboratory of Excellence GR-Ex; Paris France
- Unité des Maladies Génétiques du Globule Rouge; Hôpital Universitaire Henri Mondor (AP-HP); Créteil France
| | - Véronique Baudin-Creuza
- Institut National de la Santé et de la Recherche Médicale (Inserm)-U955; équipe 2 : Transfusion et Maladies du Globule Rouge; Institut Mondor de Recherche Biomédicale (IMRB); Université de Paris Est Créteil (UPEC); Créteil France
- Laboratory of Excellence GR-Ex; Paris France
| |
Collapse
|
99
|
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: 113] [Impact Index Per Article: 16.1] [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
|
100
|
Satta S, Paglietti ME, Sollaino MC, Barella S, Moi P, Desogus MF, Demartis FR, Manunza L, Origa R. Changes in HbA2 and HbF in alpha thalassemia carriers with KLF1 mutation. Blood Cells Mol Dis 2017; 64:30-32. [DOI: 10.1016/j.bcmd.2017.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 12/29/2022]
|