Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Int J Mol Sci ; 25(6)2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38542374

ABSTRACT

In this short review, we presented and discussed studies on the expression of globin genes in ß-thalassemia, focusing on the impact of α-globin gene expression and α-globin modifiers on the phenotype and clinical severity of ß-thalassemia. We first discussed the impact of the excess of free α-globin on the phenotype of ß-thalassemia. We then reviewed studies focusing on the expression of α-globin-stabilizing protein (AHSP), as a potential strategy of counteracting the effects of the excess of free α-globin on erythroid cells. Alternative processes controlling α-globin excess were also considered, including the activation of autophagy by ß-thalassemia erythroid cells. Altogether, the studies reviewed herein are expected to have a potential impact on the management of patients with ß-thalassemia and other hemoglobinopathies for which reduction in α-globin excess is clinically beneficial.


Subject(s)
Hemoglobinopathies , beta-Thalassemia , Humans , beta-Thalassemia/genetics , alpha-Globins/genetics , alpha-Globins/metabolism , Hemoglobinopathies/genetics , Phenotype , Gene Expression , Blood Proteins/genetics , Molecular Chaperones/genetics
2.
Int J Mol Sci ; 24(20)2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37894732

ABSTRACT

The ß-thalassemias are hereditary monogenic diseases characterized by a low or absent production of adult hemoglobin and excess in the content of α-globin. This excess is cytotoxic for the erythroid cells and responsible for the ß-thalassemia-associated ineffective erythropoiesis. Therefore, the decrease in excess α-globin is a relevant clinical effect for these patients and can be realized through the induction of fetal hemoglobin, autophagy, or both. The in vivo effects of sirolimus (rapamycin) and analogs on the induction of fetal hemoglobin (HbF) are of key importance for therapeutic protocols in a variety of hemoglobinopathies, including ß-thalassemias. In this research communication, we report data showing that a decrease in autophagy-associated p62 protein, increased expression of ULK-1, and reduction in excess α-globin are occurring in erythroid precursors (ErPCs) stimulated in vitro with low dosages of sirolimus. In addition, increased ULK-1 mRNA content and a decrease in α-globin content were found in ErPCs isolated from ß-thalassemia patients recruited for the NCT03877809 clinical trial and treated with 0.5-2 mg/day sirolimus. Our data support the concept that autophagy, ULK1 expression, and α-globin chain reduction should be considered important endpoints in sirolimus-based clinical trials for ß-thalassemias.


Subject(s)
beta-Thalassemia , Adult , Humans , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Fetal Hemoglobin , alpha-Globins/genetics , alpha-Globins/metabolism , RNA, Messenger/genetics , Autophagy , Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins/genetics
3.
Nat Commun ; 13(1): 6641, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333299

ABSTRACT

Determining the mechanisms by which genes are switched on and off during development is a key aim of current biomedical research. Gene transcription has been widely observed to occur in a discontinuous fashion, with short bursts of activity interspersed with periods of inactivity. It is currently not known if or how this dynamic behaviour changes as mammalian cells differentiate. To investigate this, using an on-microscope analysis, we monitored mouse α-globin transcription in live cells throughout erythropoiesis. We find that changes in the overall levels of α-globin transcription are most closely associated with changes in the fraction of time a gene spends in the active transcriptional state. We identify differences in the patterns of transcriptional bursting throughout differentiation, with maximal transcriptional activity occurring in the mid-phase of differentiation. Early in differentiation, we observe increased fluctuation in transcriptional activity whereas at the peak of gene expression, in early erythroblasts, transcription is relatively stable. Later during differentiation as α-globin expression declines, we again observe more variability in transcription within individual cells. We propose that the observed changes in transcriptional behaviour may reflect changes in the stability of active transcriptional compartments as gene expression is regulated during differentiation.


Subject(s)
Erythroblasts , Erythropoiesis , Mice , Animals , Erythroblasts/metabolism , Cell Differentiation/genetics , Erythropoiesis/genetics , Chromatin/metabolism , alpha-Globins/genetics , alpha-Globins/metabolism , Transcription, Genetic , Globins/genetics , Mammals/genetics
4.
Mol Biol Cell ; 33(12): br21, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35976696

ABSTRACT

It is generally believed that human mature erythrocytes do not possess functional ribosomes and therefore cannot synthesize proteins. However, the absence of translation is not consistent with the long lifespan of mature erythrocytes. They stay viable and functional for about 115 d in the circulatory system. Here, using a highly pure preparation of human mature erythrocytes, we demonstrate the presence of translation by polysome profiling, [35S]methionine labeling, and RiboPuromycylation. [35S]methionine labeling revealed that the translation in mature erythrocytes is about 10% of that observed in reticulocytes. We could observe polysomes by transmission electron microscopy in these cells. RNA-seq and quantitative real-time PCR performed on polysome fractions of these cells revealed that HBA (α-globin) and HBB (ß-globin) transcripts are translated. Using a luciferase-based reporter assay and mutational studies, we show that the sequence of the 5' untranslated region is crucial for the translation of these transcripts. Furthermore, mature erythrocytes showed reduced expression of globin proteins (α- and ß-) when treated with translation inhibitors. Overall, we provide multiple lines of evidence for translation of globin mRNAs in human mature erythrocytes.


Subject(s)
Erythrocytes , beta-Globins , 5' Untranslated Regions , Erythrocytes/metabolism , Humans , Methionine/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism
5.
Exp Hematol ; 112-113: 9-14.e7, 2022.
Article in English | MEDLINE | ID: mdl-35839944

ABSTRACT

The accumulation of unbound α-globin chains in red blood cells is a crucial pathophysiology of ß-thalassemia. IOX1 (5-carboxy-8-hydroxyquinoline) is a broad-spectrum 2-oxoglutarate (2OG)-dependent oxygenase inhibitor that can reduce α-globin mRNA expression in human cord blood erythroid progenitor cells. Therefore, IOX1 has been proposed as a potential compound for ß-thalassemia treatment through the decrease in α-globin chain synthesis. However, there is no empirical evidence regarding the consequences of IOX1 in ß-thalassemia. In this study, the therapeutic effects of IOX1 were investigated in ß0-thalassemia/hemoglobin E (HbE) erythroid progenitor cells during in vitro erythropoiesis. The results indicated that IOX1 had no impact on α-globin gene expression, but it led instead to significant decreases in γ-globin and fetal hemoglobin (HbF, α2γ2) production without affecting well-known globin regulators: KLF1, BCL11A, LRF, and GATA1. In addition, differential mRNA expression of several genes in the hypoxia response pathway revealed the induction of EGLN1, the PHD2-encoding gene, as a result of IOX1 treatment. These findings suggested that IOX1 fails to lower α-globin gene expression; on the contrary, it mediates γ-globin and HbF silencing in ß0-thalassemia/HbE erythroid progenitor cells. Because of the negative correlation of EGLN1 and γ-globin gene expression after IOX1 treatment, repurposing IOX1 to study the hypoxia response pathway and γ-globin regulation may provide beneficial information for ß-thalassemia.


Subject(s)
Hemoglobin E , Thalassemia , beta-Thalassemia , Adult , Carrier Proteins/metabolism , Erythroid Cells/metabolism , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin , Hemoglobin E/genetics , Hemoglobin E/metabolism , Humans , Hypoxia/metabolism , RNA, Messenger/genetics , Thalassemia/metabolism , alpha-Globins/metabolism , beta-Thalassemia/therapy , gamma-Globins/genetics
6.
J Trauma Acute Care Surg ; 92(1): 159-166, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34538821

ABSTRACT

BACKGROUND: Severe injury predisposes patients to trauma-induced coagulopathy, which may be subdivided by the state of fibrinolysis. Systemic hyperfibrinolysis (HF) occurs in approximately 25% of these patients with mortality as high as 70%. Severe injury also causes the release of numerous intracellular proteins, which may affect coagulation, one of which is hemoglobin, and hemoglobin substitutes induce HF in vitro. We hypothesize that the α-globin chain of hemoglobin potentiates HF in vitro by augmenting plasmin activity. METHODS: Proteomic analysis was completed on a pilot study of 30 injured patients before blood component resuscitation, stratified by their state of fibrinolysis, plus 10 healthy controls. Different concentrations of intact hemoglobin A, the α- and ß-globin chains, or normal saline (controls) were added to whole blood, and tissue plasminogen activator (tPA)-challenged thrombelastography was used to assess the degree of fibrinolysis. Interactions with plasminogen (PLG) were evaluated using surface plasmon resonance. Tissue plasminogen activator-induced plasmin activity was evaluated in the presence of the α-globin chain. RESULTS: Only the α- and ß-globin chains increased in HF patients (p < 0.01). The α-globin chain but not hemoglobin A or the ß-globin chain decreased the reaction time and significantly increased lysis time 30 on citrated native thrombelastographies (p < 0.05). The PLG and α-globin chain had interaction kinetics similar to tPA:PLG, and the α-globin chain increased tPA-induced plasmin activity. CONCLUSIONS: The α-globin chain caused HF in vitro by binding to PLG and augmenting plasmin activity and may represent a circulating "moonlighting" mediator released by the tissue damage and hemorrhagic shock inherent to severe injury. LEVEL OF EVIDENCE: Prognostic, level III.


Subject(s)
Blood Coagulation Disorders , Fibrinolysin/metabolism , Fibrinolysis , Tissue Plasminogen Activator/pharmacology , Wounds and Injuries , beta-Globins/metabolism , Adult , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/etiology , Female , Fibrinolysis/drug effects , Fibrinolysis/physiology , Fibrinolytic Agents/pharmacology , Hemoglobins/metabolism , Humans , Male , Metabolic Networks and Pathways , Prognosis , Proteomics/methods , Thrombelastography/methods , Wounds and Injuries/blood , Wounds and Injuries/complications , alpha-Globins/metabolism
7.
Am J Hematol ; 96(11): 1518-1531, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34347889

ABSTRACT

The treatment landscape for patients with ß-thalassemia is witnessing a swift evolution, yet several unmet needs continue to persist. Patients with transfusion-dependent ß-thalassemia (TDT) primarily rely on regular transfusion and iron chelation therapy, which can be associated with considerable treatment burden and cost. Patients with non-transfusion-dependent ß-thalassemia (NTDT) are also at risk of significant morbidity due to the underlying anemia and iron overload, but treatment options in this patient subgroup are limited. In this review, we provide updates on clinical trials of novel therapies targeting the underlying pathology in ß-thalassemia, including the α/non-α-globin chain imbalance, ineffective erythropoiesis, and iron dysregulation.


Subject(s)
beta-Thalassemia/therapy , Blood Transfusion , Clinical Trials as Topic , Drug Discovery , Erythropoiesis/drug effects , Humans , Iron/metabolism , Iron Chelating Agents/therapeutic use , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
8.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806017

ABSTRACT

Preeclampsia (PE) is a pregnancy disorder associated with placental dysfunction and elevated fetal hemoglobin (HbF). Early in pregnancy the placenta harbors hematopoietic stem and progenitor cells (HSPCs) and is an extramedullary source of erythropoiesis. However, globin expression is not unique to erythroid cells and can be triggered by hypoxia. To investigate the role of the placenta in increasing globin levels previously reported in PE, flow cytometry, histological and immunostaining and in situ analyses were used on placenta samples and ex vivo explant cultures. Our results indicated that in PE pregnancies, placental HSPC homing and erythropoiesis were not affected. Non-erythroid alpha-globin mRNA and protein, but not gamma-globin, were detected in syncytiotrophoblasts and stroma of PE placenta samples. Similarly, alpha-globin protein and mRNA were upregulated in normal placenta explants cultured in hypoxia. The upregulation was independent of HIF1 and NRF2, the two main candidates of globin transcription in non-erythroid cells. Our study is the first to demonstrate alpha-globin mRNA expression in syncytiotrophoblasts in PE, induced by hypoxia. However, gamma-globin was only expressed in erythrocytes. We conclude that alpha-globin, but not HbF, is expressed in placental syncytiotrophoblasts in PE and may contribute to the pathology of the disease.


Subject(s)
Hypoxia/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , alpha-Globins/metabolism , Antigens, CD34/metabolism , Biopsy , Erythroid Cells/metabolism , Erythropoiesis , Female , Flow Cytometry , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Situ Hybridization , NF-E2-Related Factor 2/metabolism , Pregnancy , RNA, Messenger/metabolism , Up-Regulation , gamma-Globins/metabolism
9.
Sci Rep ; 11(1): 8552, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879818

ABSTRACT

The reactivation of γ-globin chain synthesis to combine with excess free α-globin chains and form fetal hemoglobin (HbF) is an important alternative treatment for ß-thalassemia. We had reported HbF induction property of natural curcuminoids, curcumin (Cur), demethoxycurcumin (DMC) and bis-demethoxycurcumin (BDMC), in erythroid progenitors. Herein, the HbF induction property of trienone analogs of the three curcuminoids in erythroleukemic K562 cell lines and primary human erythroid progenitor cells from ß-thalassemia/HbE patients was examined. All three trienone analogs could induce HbF synthesis. The most potent HbF inducer in K562 cells was trienone analog of BDMC (T-BDMC) with 2.4 ± 0.2 fold increase. In addition, DNA methylation at CpG - 53, - 50 and + 6 of Gγ-globin gene promoter in K562 cells treated with the compounds including T-BDMC (9.3 ± 1.7%, 7.3 ± 1.7% and 5.3 ± 0.5%, respectively) was significantly lower than those obtained from the control cells (30.7 ± 3.8%, 25.0 ± 2.9% and 7.7 ± 0.9%, respectively P < 0.05). The trienone compounds also significantly induced HbF synthesis in ß-thalassemia/HbE erythroid progenitor cells with significantly reduction in DNA methylation at CpG + 6 of Gγ-globin gene promoter. These results suggested that the curcuminoids and their three trienone analogs induced HbF synthesis by decreased DNA methylation at Gγ-globin promoter region, without effect on Aγ-globin promoter region.


Subject(s)
Diarylheptanoids/pharmacology , Fetal Hemoglobin/biosynthesis , alpha-Globins/metabolism , beta-Thalassemia/drug therapy , gamma-Globins/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Demethylation , Diarylheptanoids/analogs & derivatives , Erythroid Precursor Cells/metabolism , Humans , Promoter Regions, Genetic , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology , gamma-Globins/chemistry , gamma-Globins/metabolism
11.
Front Immunol ; 11: 561947, 2020.
Article in English | MEDLINE | ID: mdl-33178189

ABSTRACT

Sickle cell disease (SCD) is a hemoglobinopathy affecting multiple organs and featuring acute and chronic pain. Purkinje cell damage and hyperalgesia have been demonstrated in transgenic sickle mice. Purkinje cells are associated with movement and neural function which may influence pain. We hypothesized that Purkinje cell damage and/or chronic pain burden provoke compensatory gait changes in sickle mice. We found that Purkinje cells undergoe increased apoptosis as shown by caspase-3 activation. Using an automated gait measurement system, MouseWalker, we characterized spatiotemporal gait characteristics of humanized transgenic BERK sickle mice in comparison to control mice. Sickle mice showed alteration in stance instability and dynamic gait parameters (walking speed, stance duration, swing duration and specific swing indices). Differences in stance instability may reflect motor dysfunction due to damaged Purkinje cells. Alterations in diagonal and all stance indices indicative of hesitation during walking may originate from motor dysfunction and/or arise from fear and/or anticipation of movement-evoked pain. We also demonstrate that stance duration, diagonal swing indices and all stance indices correlate with both mechanical and deep tissue hyperalgesia, while stance instability correlates with only deep tissue hyperalgesia. Therefore, objective analysis of gait in SCD may provide insights into neurological impairment and pain states.


Subject(s)
Anemia, Sickle Cell/physiopathology , Gait/genetics , Anemia, Sickle Cell/complications , Animals , Apoptosis/genetics , Brain/pathology , Caspase 3/metabolism , Chronic Pain/complications , Disease Models, Animal , Gene Knockout Techniques , Humans , Hyperalgesia/complications , Mice , Mice, Transgenic , Phenotype , Purkinje Cells/metabolism , Purkinje Cells/pathology , Walking , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism
12.
Exp Hematol ; 89: 1-12, 2020 09.
Article in English | MEDLINE | ID: mdl-32798645

ABSTRACT

I am deeply honored to receive the International Society for Experimental Hematology (ISEH) 2020 Donald Metcalf Lecture Award. Although I am not a physician and have had no formal training in hematology, I have had the privilege of working with some of the top hematologists in the world, beginning in 1970 when Dr. David Nathan was a sabbatical visitor in my laboratory and introduced me to hematological diseases.  And I take this award to be given not just to me but to an exceptional group of MD and PhD trainees and visitors in my laboratory who have cloned and characterized many proteins and RNAs important for red cell development and function. Many of these projects involved taking exceptionally large risks in developing and employing novel experimental technologies. Unsurprisingly, all of these trainees have gone on to become leaders in hematology and, more broadly, in molecular cell biology and molecular medicine. To illustrate some of the challenges we have faced and the technologies we had to develop, I have chosen several of our multiyear projects to describe in some detail: elucidating the regulation of translation of α- and ß-globin mRNAs and the defect in beta thalassemia in the 1970s; cloning the Epo receptor and several red cell membrane proteins in the 1980s and 1990s; and more recently, determining the function of many microRNAs and long noncoding RNAs in red cell development. I summarize how we are currently utilizing single-cell transcriptomics (scRNAseq) to understand how dividing transit-amplifying burst-forming unit erythroid progenitors balance the need for more progenitor cells with the need for terminally differentiated erythroid cells, and to identify drugs potentially useful in treating Epo-resistant anemias such as Diamond Blackfan anemia. I hope that the lessons I learned in managing these diverse fellows and projects, initially without having grants to support them, will be helpful to others who would like to undertake ambitious and important lines of research in hematology.


Subject(s)
Erythroid Precursor Cells/metabolism , Hematology/history , Molecular Biology/history , Receptors, Erythropoietin/history , beta-Thalassemia/genetics , Cloning, Molecular , Erythrocytes/metabolism , Erythrocytes/pathology , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Gene Expression , History, 20th Century , History, 21st Century , Humans , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
14.
Nat Commun ; 11(1): 3778, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728076

ABSTRACT

Targeted genome editing has a great therapeutic potential to treat disorders that require protein replacement therapy. To develop a platform independent of specific patient mutations, therapeutic transgenes can be inserted in a safe and highly transcribed locus to maximize protein expression. Here, we describe an ex vivo editing approach to achieve efficient gene targeting in human hematopoietic stem/progenitor cells (HSPCs) and robust expression of clinically relevant proteins by the erythroid lineage. Using CRISPR-Cas9, we integrate different transgenes under the transcriptional control of the endogenous α-globin promoter, recapitulating its high and erythroid-specific expression. Erythroblasts derived from targeted HSPCs secrete different therapeutic proteins, which retain enzymatic activity and cross-correct patients' cells. Moreover, modified HSPCs maintain long-term repopulation and multilineage differentiation potential in transplanted mice. Overall, we establish a safe and versatile CRISPR-Cas9-based HSPC platform for different therapeutic applications, including hemophilia and inherited metabolic disorders.


Subject(s)
Cell Engineering/methods , Gene Editing , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line , Female , Gene Expression Regulation , Hemophilia A/therapy , Humans , Metabolic Diseases/therapy , Mice , Promoter Regions, Genetic/genetics , Transplantation, Autologous/methods , Transplantation, Heterologous , alpha-Globins/genetics , alpha-Globins/metabolism
15.
Pediatr Hematol Oncol ; 37(4): 326-336, 2020 May.
Article in English | MEDLINE | ID: mdl-32072846

ABSTRACT

Microcytic anemia in children is commonly attributed to iron deficiency without attempting to find the cause. Inadequate investigations to exclude hemoglobinopathies lead to missed opportunities for identification of thalassemia carriers. Here we aim to describe the relative contribution of iron deficiency and thalassemia to microcytic anemia in children. This hospital-based prospective study was conducted at the Colombo North Teaching Hospital, Ragama, Sri Lanka. All newly diagnosed patients with microcytic anemia were recruited and data were collected using an interviewer-administered questionnaire. Full blood count, blood film, serum ferritin, c-reactive protein, quantification of hemoglobin sub-types and α-globin genotype were performed using 4 ml of venous blood. A total of 104 children (Male- 60.5%) were recruited. Iron deficiency was the cause for anemia in 49% whilst 16% and 10% had α- and ß-thalassemia trait respectively. Seven (6.7%) children had co-existing iron deficiency and thalassemia trait while two coinherited α- and ß-thalassemia trait. Children with ß-thalassemia trait had significantly higher red cell count and lower mean corpuscular volume compared to children with iron deficiency. However, none of the red cell parameters were significantly different between children with α-thalassemia trait and iron deficiency. Iron deficiency contributes only to half of children with microcytic anemia; one-fourth had thalassemia trait. Co-existence of iron deficiency and thalassemia trait or co-inheritance of α- and ß-thalassemia trait were found in 9%. Parallel investigation of children with microcytic anemia to diagnose iron deficiency and thalassemia provides an opportunity to identify thalassemia carriers which is beneficial for thalassemia prevention.


Subject(s)
Anemia, Iron-Deficiency , Developing Countries , alpha-Thalassemia , beta-Thalassemia , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/prevention & control , Blood Cell Count , Child, Preschool , Female , Humans , Infant , Male , Prospective Studies , Sri Lanka , alpha-Globins/metabolism , alpha-Thalassemia/blood , alpha-Thalassemia/epidemiology , alpha-Thalassemia/prevention & control , beta-Thalassemia/blood , beta-Thalassemia/epidemiology , beta-Thalassemia/prevention & control
16.
J Clin Pathol ; 73(1): 14-16, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31434698

ABSTRACT

AIMS: Untranslated regions (UTRs) play an important role in post-transcriptional regulation of gene expression, including by modulating messenger RNA (mRNA) transport out of the nucleus, translation efficiency, subcellular localisation and stability. Any mutation in this region could alter the stability of mRNA and thereby affect protein synthesis. We analysed if a mutation located in the α complex protected region of the α1 globin gene could cause non-deletional α-thalassaemia by affecting post-transcriptional stability (mRNA stability). METHODS: A total of 14 patients without anaemia, normal or slight microcytosis and hypochromia (medium concentration haemoglobin [MCH] <27 pg) were studied. Haemoglobin subtypes were screened using capillary zone electrophoresis and ion-exchange high-performance liquid chromatography (VARIANT II ß-Thalassaemia Short Program). The most common α-globin mutations were identified by multiplex PCR (Alpha-Globin StripAssay kit) and the molecular characterisation by automatic sequencing of alpha globin genes. RESULTS: All of them shown a novel transversion mutation in nt 778 (C>A), which is located in the 3' UTR in the α complex protected region [HBA1: c.*+46C>A]. CONCLUSIONS: This mutation is in the αRNAmin binding site, so a single nucleotide substitution in this region can decrease mRNA stability by potentially compromising the binding of α-complex protein to αRNAmin, favouring the decay of α-globin mRNA via erythroid cell-enriched endoribonuclease cleavage. In this case, it is a non-deletional α-thalassaemia. However, in silico and empirical studies predicted that it could be a silent polymorphism. Functional studies should be carried out to confirm whether it is a pathological mutation or a silent polymorphism.


Subject(s)
3' Untranslated Regions , Mutation , Polymorphism, Genetic , RNA Stability , RNA, Messenger/genetics , alpha-Globins/genetics , alpha-Thalassemia/genetics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Multiplex Polymerase Chain Reaction , Phenotype , RNA, Messenger/metabolism , Risk Factors , alpha-Globins/metabolism , alpha-Thalassemia/blood , alpha-Thalassemia/diagnosis
17.
Mol Cell Neurosci ; 100: 103398, 2019 10.
Article in English | MEDLINE | ID: mdl-31472221

ABSTRACT

Neurogenesis is driven by spatially and temporally regulated proliferation of neuronal progenitor cells that generates enormous number of assorted neurons to drive the complex behavior of an organism. Drosophila nervous system provides an advantageous model for identification and elucidation of the functional significance of the novel gene(s) involved in neurogenesis. The present study attempts to investigate the role(s) of globin1 (glob1) in the development and maintenance of the nervous system in Drosophila. It is increasingly clear now that globin genes play important role(s) in the various biological phenomena. The vertebrate neuroglobin has been reported to profoundly express in neuronal tissues and provides neuroprotection. We noted ubiquitous presence of Glob1 in the developing neuronal tissues with enhanced concentration throughout the VNC which comprises of midline cell clusters, which subsequently forms numerous types of progenitor cells and finally differentiate into specific neurons of the nervous system. Ubiquitous or pan-neuronal downregulation of glob1 causes partial lethality and mis-positioning of various neural-progenitor cells present in the embryonic midline cell clusters. Subsequently, profound expression of Glob1 was noted in the outer proliferation center of larval brain and photoreceptor axons of optic stalk. The overall arrangement of photoreceptor axons and stereotype positioning of neuroblast cells present in the central region of the brain were severally affected due to reduced expression of glob1. In addition, such larvae and surviving adults develop significant neuro-muscular disabilities. For the first time, our study suggests a novel role of glob1 in development and maintenance of the nervous system adding a new dimension to the functional significance of the multi-tasking glob1 gene in Drosophila.


Subject(s)
Drosophila Proteins/genetics , Ganglia, Invertebrate/metabolism , Neurogenesis , alpha-Globins/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/embryology , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , alpha-Globins/metabolism
18.
Sci Transl Med ; 11(506)2019 08 21.
Article in English | MEDLINE | ID: mdl-31434755

ABSTRACT

In ß-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate ß-thalassemia pathophysiology by degrading toxic free α-globin, although the associated mechanisms are poorly understood. We show that loss of the autophagy-activating Unc-51-like kinase 1 (Ulk1) gene in ß-thalassemic mice reduces autophagic clearance of α-globin in red blood cell precursors and exacerbates disease phenotypes, whereas inactivation of the canonical autophagy-related 5 (Atg5) gene has relatively minor effects. Systemic treatment with the mTORC1 inhibitor rapamycin reduces α-globin precipitates and lessens pathologies in ß-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of ß-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating ß-thalassemia.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , alpha-Globins/metabolism , beta-Thalassemia/enzymology , beta-Thalassemia/pathology , Animals , Antigens, CD34/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , Disease Progression , Enzyme Activation/drug effects , Gene Deletion , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Phenotype , Reticulocytes/drug effects , Reticulocytes/metabolism , Reticulocytes/ultrastructure , Sirolimus/pharmacology
19.
Clin Chim Acta ; 498: 154-160, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31449773

ABSTRACT

BACKGROUND: Glycated hemoglobin (Hemoglobin A1c, HbA1c) plays a key role in monitoring long-term blood glucose levels in diabetics mellitus. Therefore, it is of great importance to ensure test quality of HbA1c methods. OBJECTIVES: We aimed to evaluate analytical performances of a matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS) system for the measurement of HbA1c. METHODS: We assessed the analytical performances of the method including imprecision, accuracy, and linearity. In addition, comparison with Variant II Turbo 2.0 and Capillarys3 TERA, correlation between glycation rate of α and ß globin as well as the influence of most frequent analytical interferences in HbA1c assays were also investigated. RESULTS: As measurement of imprecision, within-run CVs and total CVs were lower than 1.6% and 2.4%, respectively. Discrepancy of test results (<0.2%) of IFCC value-assigned external quality control samples indicated a good accuracy of the method. The linearity was excellent with a correlation coefficient of 0.999. The QuanTOF results were well correlated with those obtained by Variant II Turbo 2.0 and Capillarys3 TERA. Good correlation between glycation rates of α and ß globin were found. QuanTOF was not prone to common interferences including bilirubin, triglyceride, labile A1c, and carbamylated hemoglobin. However, unacceptable positive bias was observed when the amount of HbF were greater than approximately 8.0% or in the presence of HbS. CONCLUSIONS: QuanTOF perform well for the determination of HbA1c and meet quality criteria requested for clinical use.


Subject(s)
Glycated Hemoglobin/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Bias , Glycosylation , Kinetics , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/standards , alpha-Globins/metabolism , beta-Globins/metabolism
20.
Mol Biol Rep ; 46(5): 5041-5048, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31273613

ABSTRACT

Thalassemia is one of the most common monogenic hereditary disorders. Despite noticeable advances made in prevention strategies, it is still highly prevalent in the Iranian population. A key approach to management and early diagnosis of the disease is through revealing the regions with high prevalence and determining common genetic and phenotypic diversity. In the current study Hemoglobin H (HbH) disease patients were analyzed as the most common form of thalassemia intermedia in Iran. A total of 80 patients suspected of being thalassemic according to their mild to moderate anemia, microcytosis and normal iron levels were included in this study at the hemoglobinopathy and thalassemia center of Ahvaz University of Medical Science. Patients were analyzed for hematological parameters and HbH mutations using Multiplex Gap Polymerase Chain Reaction and Multiplex Amplification Refractory Mutation System. Twelve mutations were detected in the studied population. The most common genotype was -α3.7/--MED (45%) followed by Homozygote αPoly A2 (17.5%). A total of ten different alpha-globin (α-globin) mutations were observed in patients which --MED, being the most common mutation (26.27%), followed by -α3.7 (24.37%) and αpolyA2(A>G) (18.12%). Hematological parameters such as Hb, MCV, MCH and HbH were assessed and results showed that they varied significantly among genotypes, adjusted to age and gender. This study reveals a highly diverse range of HbH patients different from what was thought in terms of both genotype and phenotype in the Khuzestan region of Iran. These findings could contribute to improve the thalassemia managing policies in this province.


Subject(s)
Thalassemia/genetics , alpha-Thalassemia/genetics , Adolescent , Adult , Female , Genetic Association Studies/methods , Genotype , Humans , Iran/epidemiology , Male , Mutation , Phenotype , Thalassemia/metabolism , Young Adult , alpha-Globins/genetics , alpha-Globins/metabolism , alpha-Thalassemia/metabolism , beta-Thalassemia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...