1
|
Theocharaki K, Anastasiadi AT, Delicou S, Tzounakas VL, Barla I, Rouvela S, Kazolia E, Tzafa G, Mpekoulis G, Gousdovas T, Pavlou E, Kostopoulos IV, Velentzas AD, Simantiris N, Xydaki A, Vassilaki N, Voskaridou E, Aggeli IK, Nomikou E, Tsitsilonis O, Papageorgiou E, Thomaidis N, Gikas E, Politou M, Komninaka V, Antonelou MH. Cellular and biochemical heterogeneity contributes to the phenotypic diversity of transfusion-dependent β-thalassemia. Blood Adv 2025; 9:2091-2107. [PMID: 39928952 PMCID: PMC12051129 DOI: 10.1182/bloodadvances.2024015232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
ABSTRACT Transfusion-dependent thalassemia (TDT) is a type of protein aggregation disease. Its clinical heterogeneity imposes challenges in effective management. Red blood cell (RBC) variables may be clinically relevant as mechanistic parts or tellers of TDT pathophysiology. This is a cross-sectional study of RBC and plasma physiology in adult patients with TDT vs healthy control. TDT plasma was characterized by increased protein carbonylation, antioxidants, and larger than normal extracellular vesicles. RBCs were osmotically resistant but prone to oxidative hemolysis. They overexposed phosphatidylserine and exhibited pathologically low proteasome proteolytic activity (PPA), which correlated with metabolic markers of the disease. RBC ultrastructure was distorted, with splenectomy-related membrane pits of 300 to 800 nm. Plasma metabolomics revealed differences in heme metabolism, redox potential, short-chain fatty acids, and nitric oxide bioavailability, but also in catecholamine pathways. According to coefficient of variation assessment, hemolysis, iron homeostasis, PPA, and phosphatidylserine exposure were highly variable among patients, as opposed to RBC fragility and plasma antioxidants, amino acids, and catecholamines. Sex-based differences were detected in hemolysis, redox, and energy variables, whereas splenectomy-related differences referred to thrombotic risk, RBC morphology, and plasma metabolites with neuroendocrine activity. Hepcidin varied according to oxidative hemolysis and metabolic markers of bacterial activity. Patients with higher pretransfusion hemoglobin levels (>10 g/dL) presented mildly distorted profiles and lower membrane-associated PPA, whereas classification by severity of mutations revealed different levels of hemostasis, inflammation, plasma epinephrine, hexosamines, and methyltransferase activity markers. The currently reported heterogeneity of cellular and biochemical features probably contributes to the wide phenotypic diversity of TDT at clinical level.
Collapse
Affiliation(s)
- Konstantina Theocharaki
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Alkmini T. Anastasiadi
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Delicou
- Thalassemia and Sickle Cell Unit, Expertise Center of Hemoglobinopathies and their Complications, Hippokration General Hospital of Athens, Athens, Greece
| | - Vassilis L. Tzounakas
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Barla
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Stella Rouvela
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evgenia Kazolia
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Tzafa
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - George Mpekoulis
- Department of Microbiology, Laboratory of Molecular Virology, Hellenic Pasteur Institute, Athens, Greece
| | - Theodore Gousdovas
- Hematology Laboratory-Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymia Pavlou
- Blood Bank and Haemophilia Unit, Hippokration General Hospital of Athens, Athens, Greece
| | - Ioannis V. Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanassios D. Velentzas
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Aikaterini Xydaki
- Thalassemia and Sickle Cell Unit, Expertise Center of Hemoglobinopathies and their Complications, Hippokration General Hospital of Athens, Athens, Greece
| | - Niki Vassilaki
- Department of Microbiology, Laboratory of Molecular Virology, Hellenic Pasteur Institute, Athens, Greece
| | - Ersi Voskaridou
- Centre of Excellence in Rare Haematological (Haemoglobinopathies) & Rare Metabolic (Gaucher Disease) Diseases, Laiko General Hospital, Athens, Greece
| | - Ioanna-Katerina Aggeli
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Efrosyni Nomikou
- Blood Bank and Haemophilia Unit, Hippokration General Hospital of Athens, Athens, Greece
| | - Ourania Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathia Papageorgiou
- Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica, Egaleo, Greece
| | - Nikolaos Thomaidis
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Gikas
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna Politou
- Hematology Laboratory-Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Veroniki Komninaka
- Centre of Excellence in Rare Haematological (Haemoglobinopathies) & Rare Metabolic (Gaucher Disease) Diseases, Laiko General Hospital, Athens, Greece
| | - Marianna H. Antonelou
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
2
|
Long Y, Zhang Q, Ling L, Zhuang Y, Wei X, Huang H, Lu Z, Huang Y, Chen X, Ye Y, Feng X, Zhang H, Huang B, Huang Y, Liang Y, Fang M, Nakamura Y, Lin B, Zhang X, Lu D, Jin X, Xu X. Mutations in AMBRA1 aggravate β-thalassemia by impairing autophagy-mediated clearance of free α-globin. Blood 2025; 145:1074-1088. [PMID: 39693613 DOI: 10.1182/blood.2023022688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 12/20/2024] Open
Abstract
ABSTRACT Accumulation of free α-globin is a critical factor in the pathogenesis of β-thalassemia. Autophagy plays a crucial role in clearing toxic free α-globin, thereby reducing disease severity. However, the impact of natural mutations in autophagy-related genes (ATGs) on the phenotypic variability of β-thalassemia remains unclear. In this study, we systematically investigated the relationship between variants in ATGs and disease phenotypes in a cohort of 1022 patients with β-thalassemia, identifying 4 missense mutations in the autophagy and beclin 1 regulator 1 (AMBRA1) gene. Disruption of the Ambra1 gene in β-thalassemic mice was found to reduce autophagic clearance of α-globin in red blood cell precursors, exacerbating disease phenotypes. Functional characterization of the AMBRA1 gene and these mutations in patient-derived CD34+ cells, edited human umbilical cord blood-derived erythroid progenitor 2 (HUDEP-2) cells, and engineered HUDEP-2 β-thalassemic cells confirmed that AMBRA1 facilitates the autophagic clearance of free α-globin in human erythroid cells. Functional studies demonstrated that AMBRA1 missense mutants destabilize Unc-51-like kinase 1 protein, inhibit light chain 3 protein lipidation, and subsequently hinder autophagic flux, leading to increased α-globin deposition. Additionally, these mutations were associated with erythrotoxic effects in vitro, including increased intracellular reactive oxygen species levels, higher apoptosis rates, and impaired erythroid differentiation and maturation. This study sheds light on the molecular association between mutations in ATGs and the exacerbation of β-thalassemia, highlighting the potential role of the AMBRA1 gene as a promising diagnostic and therapeutic target for β-hemoglobinopathies.
Collapse
Affiliation(s)
- Yong Long
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianqian Zhang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, China
| | - Ling Ling
- Yangzhou University, Yangzhou, China
| | - Yuan Zhuang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoyang Huang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhanping Lu
- Central Laboratory, Chongqing University Fuling Hospital, Chongqing University, Chongqing, China
| | - Yushan Huang
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xianming Chen
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuhua Ye
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haokun Zhang
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Binbin Huang
- Department 1 of Internal Medicine, Sixth People's Hospital of Nanning, Nanning, China
| | - Yueyan Huang
- Department of Pediatric, Affiliated Hospital of Youjiang Medical University for Nationalities Baise, Baise, China
| | - Yidan Liang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mingyan Fang
- Central Laboratory, Chongqing University Fuling Hospital, Chongqing University, Chongqing, China
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Japan
- Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Bin Lin
- Genetics Laboratory, Guangzhou Huayin Healthcare Group Co, Ltd, Guangzhou, China
- Genetics Laboratory, Guangzhou Jiexu Gene Technology Co, Ltd, Guangzhou, China
| | - Xinhua Zhang
- Department of Hematology, 923rd Hospital of the People's Liberation Army, Nanning, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xin Jin
- Central Laboratory, Chongqing University Fuling Hospital, Chongqing University, Chongqing, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiangmin Xu
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Lechauve C, Weiss MJ. AMBRA1 performs a balancing act in β-thalassemia. Blood 2025; 145:1001-1003. [PMID: 40048224 DOI: 10.1182/blood.2024027597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025] Open
|
4
|
Pahelkar A, Sharma D, Vohra P, Sawant S. Leveraging Multi-Omics Approaches and Advanced Technologies to Unravel the Molecular Complexities, Modifiers, and Precision Medicine Strategies for Hemoglobin H Disease. Eur J Haematol 2024; 113:738-744. [PMID: 39385444 DOI: 10.1111/ejh.14319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
Hemoglobin H (HbH) disease, a form of alpha-thalassemia, poses significant clinical challenges due to its complex molecular underpinnings. It is characterized by reduced synthesis of the alpha-globin chain. The integration of multi-omics and precision medicine holds immense potential to comprehensively understand and capture interactions at the molecular and genetic levels. This review integrates current multi-omics approaches and advanced technologies in HbH research. Furthermore, it delves into detailed pathophysiology and possible therapeutics in the upcoming future. We explore the role of genomics, transcriptomics, proteomics, and metabolomics studies, alongside bioinformatics tools and gene-editing technologies like CRISPR/Cas9, to identify genetic modifiers, decipher molecular pathways, and discover therapeutic targets. Recent advancements are unveiling novel genetic and epigenetic modifiers impacting HbH disease severity, paving the way for personalized precision medicine interventions. The significance of multi-omics research in unraveling the complexities of rare diseases like HbH is underscored, highlighting its potential to revolutionize clinical practice through precision medicine approaches. This paradigm shift can pave the way for a deeper understanding of HbH complexities and improved disease management.
Collapse
Affiliation(s)
- Akshata Pahelkar
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| | - Deep Sharma
- MES's H. K. College of Pharmacy, Mumbai, Maharashtra, India
| | - Payaam Vohra
- MES's H. K. College of Pharmacy, Mumbai, Maharashtra, India
| | - Sayli Sawant
- MES's H. K. College of Pharmacy, Mumbai, Maharashtra, India
| |
Collapse
|
5
|
Abbineni PS, Baid S, Weiss MJ. A moonlighting job for α-globin in blood vessels. Blood 2024; 144:834-844. [PMID: 38848504 PMCID: PMC11830976 DOI: 10.1182/blood.2023022192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
ABSTRACT Red blood cells express high levels of hemoglobin A tetramer (α2β2) to facilitate oxygen transport. Hemoglobin subunits and related proteins are also expressed at lower levels in other tissues across the animal kingdom. Physiological functions for most nonerythroid globins likely derive from their ability to catalyze reduction-oxidation (redox) reactions via electron transfer through heme-associated iron. An interesting example is illustrated by the recent discovery that α-globin without β-globin is expressed in some arteriolar endothelial cells (ECs). α-globin binds EC nitric oxide (NO) synthase (eNOS) and degrades its enzymatic product NO, a potent vasodilator. Thus, depletion of α-globin in ECs or inhibition of its association with eNOS causes arteriolar relaxation and lowering of blood pressure in mice. Some of these findings have been replicated in isolated human blood vessels, and genetic studies are tractable in populations in which α-thalassemia alleles are prevalent. Two small studies identified associations between loss of α-globin genes in humans and NO-regulated vascular responses elicited by local hypoxia-induced blood flow or thermal stimulation. In a few larger population-based studies, no associations were detected between loss of α-globin genes and blood pressure, ischemic stroke, or pulmonary hypertension. In contrast, a significant positive association between α-globin gene copy number and kidney disease was detected in an African American cohort. Further studies are required to define comprehensively the expression of α-globin in different vascular beds and ascertain their overall impact on normal and pathological vascular physiology.
Collapse
Affiliation(s)
- Prabhodh S. Abbineni
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Srishti Baid
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Mitchell J. Weiss
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
6
|
Zhang H, Liu R, Fang Z, Nie L, Ma Y, Sun F, Mei J, Song Z, Ginzburg YZ, Liu J, Chen H. Mitoxantrone ameliorates ineffective erythropoiesis in a β-thalassemia intermedia mouse model. Blood Adv 2024; 8:4017-4024. [PMID: 38861356 PMCID: PMC11339037 DOI: 10.1182/bloodadvances.2024012679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
ABSTRACT β-thalassemia is a condition characterized by reduced or absent synthesis of β-globin resulting from genetic mutations, leading to expanded and ineffective erythropoiesis. Mitoxantrone has been widely used clinically as an antitumor agent considering its ability to inhibit cell proliferation. However, its therapeutic effect on expanded and ineffective erythropoiesis in β-thalassemia is untested. We found that mitoxantrone decreased α-globin precipitates and ameliorated anemia, splenomegaly, and ineffective erythropoiesis in the HbbTh3/+ mouse model of β-thalassemia intermedia. The partially reversed ineffective erythropoiesis is a consequence of effects on autophagy as mitochondrial retention and protein levels of mTOR, P62, and LC3 in reticulocytes decreased in mitoxantrone-treated HbbTh3/+ mice. These data provide significant preclinical evidence for targeting autophagy as a novel therapeutic approach for β-thalassemia.
Collapse
Affiliation(s)
- Haihang Zhang
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Rui Liu
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Zheng Fang
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ling Nie
- Xiangya Hospital, Central South University, Changsha, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Jingjing Mei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Zhiyin Song
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yelena Z. Ginzburg
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Huiyong Chen
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| |
Collapse
|
7
|
Gambari R, Finotti A. Therapeutic Relevance of Inducing Autophagy in β-Thalassemia. Cells 2024; 13:918. [PMID: 38891049 PMCID: PMC11171814 DOI: 10.3390/cells13110918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The β-thalassemias are inherited genetic disorders affecting the hematopoietic system. In β-thalassemias, more than 350 mutations of the adult β-globin gene cause the low or absent production of adult hemoglobin (HbA). A clinical parameter affecting the physiology of erythroid cells is the excess of free α-globin. Possible experimental strategies for a reduction in excess free α-globin chains in β-thalassemia are CRISPR-Cas9-based genome editing of the β-globin gene, forcing "de novo" HbA production and fetal hemoglobin (HbF) induction. In addition, a reduction in excess free α-globin chains in β-thalassemia can be achieved by induction of the autophagic process. This process is regulated by the Unc-51-like kinase 1 (Ulk1) gene. The interplay with the PI3K/Akt/TOR pathway, with the activity of the α-globin stabilizing protein (AHSP) and the involvement of microRNAs in autophagy and Ulk1 gene expression, is presented and discussed in the context of identifying novel biomarkers and potential therapeutic targets for β-thalassemia.
Collapse
Affiliation(s)
| | - Alessia Finotti
- Center “Chiara Gemmo and Elio Zago” for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy;
| |
Collapse
|
8
|
Oloruntola OD. Red chili powder dietary supplementation regularized the performance, hematobiochemical indices, oxidative status, and 8-hydroxy-2'-deoxyguanosine of aflatoxin B1 exposed broiler chickens. Transl Anim Sci 2024; 8:txae006. [PMID: 38313223 PMCID: PMC10836503 DOI: 10.1093/tas/txae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
The effects of red chili powder dietary supplementation on the performance, hematobiochemical indices, oxidative status, and DNA damage in broiler chickens fed aflatoxin B1 (AFB1) contaminated diets were studied. Two hundred and forty Cobb 500 breed day-old broiler chicks were randomly distributed into control group (CONT), 0.5 mg/kg AFB1-exposed group (AFTB), 0.5 g/kg red chili pericarp powder supplementation alongside the 0.5 mg/kg AFB1 exposed group (RCPA), and 0.5 g/kg red chili seed powder supplementation alongside the 0.5 mg/kg AFB1 exposed group (RCSA). The red chili supplementation, in both pericarp powder and seed powder, positively influenced broiler performance by improving (P < 0.05) weight gain, feed intake, and feed conversion ratio, with a reduction in mortality rates compared to the AFTB group. Hematological indices showed that AFB1 exposure decreased (P < 0.05) the red blood cell count, packed cell volume, and hemoglobin (Hb) concentration, but the red chili supplementation mitigated these reductions. Additionally, total white blood cell counts were maintained (P > 0.05) in red chili-supplemented groups compared to CONT. Red chili supplementation increased (P < 0.05) the total protein and globulin concentrations and reduced (P < 0.05) liver enzyme levels compared to the AFTB group. The oxidative enzyme levels in RCPA and RCSA were similar (P > 0.05) to CONT groups. The red chili supplementations counteracted DNA damage, as reflected by similar (P > 0.05) 8-hydroxy-2'-deoxyguanosine levels recorded in RCPA, RCSA, and CONT groups levels. These findings suggest that 0.5 g/kg red chili supplementation has the potential to ameliorate the adverse effects of AFB1 exposure on broiler chickens, improving their performance and health.
Collapse
|
9
|
Daniels DE, Ferrer-Vicens I, Hawksworth J, Andrienko TN, Finnie EM, Bretherton NS, Ferguson DCJ, Oliveira ASF, Szeto JYA, Wilson MC, Brewin JN, Frayne J. Human cellular model systems of β-thalassemia enable in-depth analysis of disease phenotype. Nat Commun 2023; 14:6260. [PMID: 37803026 PMCID: PMC10558456 DOI: 10.1038/s41467-023-41961-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
β-thalassemia is a prevalent genetic disorder causing severe anemia due to defective erythropoiesis, with few treatment options. Studying the underlying molecular defects is impeded by paucity of suitable patient material. In this study we create human disease cellular model systems for β-thalassemia by gene editing the erythroid line BEL-A, which accurately recapitulate the phenotype of patient erythroid cells. We also develop a high throughput compatible fluorometric-based assay for evaluating severity of disease phenotype and utilize the assay to demonstrate that the lines respond appropriately to verified reagents. We next use the lines to perform extensive analysis of the altered molecular mechanisms in β-thalassemia erythroid cells, revealing upregulation of a wide range of biological pathways and processes along with potential novel targets for therapeutic investigation. Overall, the lines provide a sustainable supply of disease cells as research tools for identifying therapeutic targets and as screening platforms for new drugs and reagents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jenn-Yeu A Szeto
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | | | - John N Brewin
- Haematology Department, King's college Hospital NHS Foundation, London, SE5 9RS, UK
- Red Cell Biology Group, Kings College London, London, SE5 9NU, UK
| | - Jan Frayne
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
10
|
Guerra A, Parhiz H, Rivella S. Novel potential therapeutics to modify iron metabolism and red cell synthesis in diseases associated with defective erythropoiesis. Haematologica 2023; 108:2582-2593. [PMID: 37345473 PMCID: PMC10542825 DOI: 10.3324/haematol.2023.283057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
Under normal conditions, iron metabolism is carefully regulated to sustain normal cellular functions and the production of hemoglobin in erythroid cells. Perturbation to the erythropoiesis-iron metabolism axis can result in iron imbalances and cause anemia or organ toxicity. Various congenital and acquired diseases associated with abnormal red cell production are characterized by aberrant iron absorption. Several recent studies have shown that improvements in red blood cell production also ameliorate iron metabolism and vice versa. Many therapeutics are now under development with the potential to improve a variety of hematologic diseases, from β-thalassemia and iron-refractory iron deficiency anemia to anemia of inflammation and polycythemia vera. This review summarizes selected mechanisms related to red cell production and iron metabolism and describes potential therapeutics and their current uses. We also consider the potential application of the discussed therapeutics on various diseases, alone or in combination. The vast repertoire of drugs under development offers new opportunities to improve the clinical care of patients suffering from congenital or acquired red blood cell disorders with limited or no treatment options.
Collapse
Affiliation(s)
- Amaliris Guerra
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA
| | - Hamideh Parhiz
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; RNA Institute, University of Pennsylvania, Philadelphia, PA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; RNA Institute, University of Pennsylvania, Philadelphia, PA, USA; Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania, Philadelphia, PA, USA; Raymond G. Perelman Center for Cellular and Molecular Therapeutics-CHOP; Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
11
|
Keith J, Christakopoulos GE, Fernandez AG, Yao Y, Zhang J, Mayberry K, Telange R, Sweileh RBA, Dudley M, Westbrook C, Sheppard H, Weiss MJ, Lechauve C. Loss of miR-144/451 alleviates β-thalassemia by stimulating ULK1-mediated autophagy of free α-globin. Blood 2023; 142:918-932. [PMID: 37339583 PMCID: PMC10517214 DOI: 10.1182/blood.2022017265] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/22/2023] Open
Abstract
Most cells can eliminate unstable or misfolded proteins through quality control mechanisms. In the inherited red blood cell disorder β-thalassemia, mutations in the β-globin gene (HBB) lead to a reduction in the corresponding protein and the accumulation of cytotoxic free α-globin, which causes maturation arrest and apoptosis of erythroid precursors and reductions in the lifespan of circulating red blood cells. We showed previously that excess α-globin is eliminated by Unc-51-like autophagy activating kinase 1 (ULK1)-dependent autophagy and that stimulating this pathway by systemic mammalian target of rapamycin complex 1 (mTORC1) inhibition alleviates β-thalassemia pathologies. We show here that disrupting the bicistronic microRNA gene miR-144/451 alleviates β-thalassemia by reducing mTORC1 activity and stimulating ULK1-mediated autophagy of free α-globin through 2 mechanisms. Loss of miR-451 upregulated its target messenger RNA, Cab39, which encodes a cofactor for LKB1, a serine-threonine kinase that phosphorylates and activates the central metabolic sensor adenosine monophosphate-activated protein kinase (AMPK). The resultant enhancement of LKB1 activity stimulated AMPK and its downstream effects, including repression of mTORC1 and direct activation of ULK1. In addition, loss of miR-144/451 inhibited the expression of erythroblast transferrin receptor 1, causing intracellular iron restriction, which has been shown to inhibit mTORC1, reduce free α-globin precipitates, and improve hematological indices in β-thalassemia. The beneficial effects of miR-144/451 loss in β-thalassemia were inhibited by the disruption of Cab39 or Ulk1 genes. Together, our findings link the severity of β-thalassemia to a highly expressed erythroid microRNA locus and a fundamental, metabolically regulated protein quality control pathway that is amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Julia Keith
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | | | - Yu Yao
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jingjing Zhang
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kalin Mayberry
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Rahul Telange
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Razan B. A. Sweileh
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Michael Dudley
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Camilla Westbrook
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Heather Sheppard
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
12
|
Zhang X, Song J, Shah BN, Han J, Hassan T, Miasniakova G, Sergueeva A, Nekhai S, Machado RF, Gladwin MT, Saraf SL, Prchal JT, Gordeuk VR. Gene expression changes in sickle cell reticulocytes and their clinical associations. Sci Rep 2023; 13:12864. [PMID: 37553354 PMCID: PMC10409856 DOI: 10.1038/s41598-023-40039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023] Open
Abstract
Transcriptional changes in compensatory erythropoiesis in sickle cell anemia (SCA) and their disease modulation are unclear. We detected 1226 differentially expressed genes in hemoglobin SS reticulocytes compared to non-anemic hemoglobin AA controls. Assessing developmental expression changes in hemoglobin AA erythroblasts for these genes suggests heightened terminal differentiation in early erythroblasts in SCA that diminishes toward the polychromatic to orthochromatic stage transition. Comparison of reticulocyte gene expression changes in SCA with that in Chuvash erythrocytosis, a non-anemic disorder of increased erythropoiesis due to constitutive activation of hypoxia inducible factors, identified 453 SCA-specific changes attributable to compensatory erythropoiesis. Peripheral blood mononuclear cells (PBMCs) in SCA contain elevated proportions of erythroid progenitors due to heightened erythropoiesis. Deconvolution analysis in PBMCs from 131 SCA patients detected 54 genes whose erythroid expression correlated with erythropoiesis efficiency, which were enriched with SCA-specific changes (OR = 2.9, P = 0.00063) and annotation keyword "ubiquitin-dependent protein catabolic process", "protein ubiquitination", and "protein polyubiquitination" (OR = 4.2, P = 7.5 × 10-5). An erythroid expression quantitative trait locus of one of these genes, LNX2 encoding an E3 ubiquitin ligase, associated with severe pain episodes in 774 SCA patients (OR = 1.7, P = 3.9 × 10-5). Thus, erythroid gene transcription responds to unique conditions within SCA erythroblasts and these changes potentially correspond to vaso-occlusive manifestations.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Jihyun Song
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Binal N Shah
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin Han
- College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Taif Hassan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | - Sergei Nekhai
- Center for Sickle Cell Disease, Howard University, Washington, DC, USA
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Santosh L Saraf
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Josef T Prchal
- Department of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Victor R Gordeuk
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Reboucas P, Fillebeen C, Botta A, Cleverdon R, Steele AP, Richard V, Zahedi RP, Borchers CH, Burelle Y, Hawke TJ, Pantopoulos K, Sweeney G. Discovery-Based Proteomics Identify Skeletal Muscle Mitochondrial Alterations as an Early Metabolic Defect in a Mouse Model of β-Thalassemia. Int J Mol Sci 2023; 24:ijms24054402. [PMID: 36901833 PMCID: PMC10002226 DOI: 10.3390/ijms24054402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Although metabolic complications are common in thalassemia patients, there is still an unmet need to better understand underlying mechanisms. We used unbiased global proteomics to reveal molecular differences between the th3/+ mouse model of thalassemia and wild-type control animals focusing on skeletal muscles at 8 weeks of age. Our data point toward a significantly impaired mitochondrial oxidative phosphorylation. Furthermore, we observed a shift from oxidative fibre types toward more glycolytic fibre types in these animals, which was further supported by larger fibre-type cross-sectional areas in the more oxidative type fibres (type I/type IIa/type IIax hybrid). We also observed an increase in capillary density in th3/+ mice, indicative of a compensatory response. Western blotting for mitochondrial oxidative phosphorylation complex proteins and PCR analysis of mitochondrial genes indicated reduced mitochondrial content in the skeletal muscle but not the hearts of th3/+ mice. The phenotypic manifestation of these alterations was a small but significant reduction in glucose handling capacity. Overall, this study identified many important alterations in the proteome of th3/+ mice, amongst which mitochondrial defects leading to skeletal muscle remodelling and metabolic dysfunction were paramount.
Collapse
Affiliation(s)
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
| | - Amy Botta
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Riley Cleverdon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Alexandra P. Steele
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Vincent Richard
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - René P. Zahedi
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Christoph H. Borchers
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Thomas J. Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +1-416-736-2100 (ext. 66635)
| |
Collapse
|
14
|
Luo P, Liu X, Tang Z, Xiong B. Decreased expression of HBA1 and HBB genes in acute myeloid leukemia patients and their inhibitory effects on growth of K562 cells. Hematology 2022; 27:1003-1009. [DOI: 10.1080/16078454.2022.2117186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Ping Luo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Xiaoyan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Zehai Tang
- Department of Emergency, The Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bei Xiong
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
15
|
Caulier A, Jankovsky N, Gautier EF, El Nemer W, Guitton C, Ouled-Haddou H, Guillonneau F, Mayeux P, Salnot V, Bruce J, Picard V, Garçon L. Red blood cell proteomics reveal remnant protein biosynthesis and folding pathways in PIEZO1-related hereditary xerocytosis. Front Physiol 2022; 13:960291. [PMID: 36531183 PMCID: PMC9751340 DOI: 10.3389/fphys.2022.960291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2023] Open
Abstract
Hereditary xerocytosis is a dominant red cell membrane disorder characterized by an increased leak of potassium from the inside to outside the red blood cell membrane, associated with loss of water leading to red cell dehydration and chronic hemolysis. 90% of cases are related to heterozygous gain of function mutations in PIEZO1, encoding a mechanotransductor that translates a mechanical stimulus into a biological signaling. Data are still required to understand better PIEZO1-HX pathophysiology. Recent studies identified proteomics as an accurate and high-input tool to study erythroid progenitors and circulating red cell physiology. Here, we isolated red blood cells from 5 controls and 5 HX patients carrying an identified and pathogenic PIEZO1 mutation and performed a comparative deep proteomic analysis. A total of 603 proteins were identified among which 56 were differentially expressed (40 over expressed and 16 under expressed) between controls and HX with a homogenous expression profile within each group. We observed relevant modifications in the protein expression profile related to PIEZO1 mutations, identifying two main "knots". The first contained both proteins of the chaperonin containing TCP1 complex involved in the assembly of unfolded proteins, and proteins involved in translation. The second contained proteins involved in ubiquitination. Deregulation of proteins involved in protein biosynthesis was also observed in in vitro-produced reticulocytes after Yoda1 exposure. Thus, our work identifies significant changes in the protein content of PIEZO1-HX erythrocytes, revealing a "PIEZO1 signature" and identifying potentially targetable pathways in this disease characterized by a heterogeneous clinical expression and contra-indication of splenectomy.
Collapse
Affiliation(s)
- Alexis Caulier
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - Nicolas Jankovsky
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - Emilie Fleur Gautier
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
- Institut Imagine-INSERM U1163, Necker Hospital, University of Paris, Paris, France
- Laboratoire d’excellence GR-Ex, Paris, France
| | | | - Corinne Guitton
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
| | - Hakim Ouled-Haddou
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - François Guillonneau
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Patrick Mayeux
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Virginie Salnot
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Johanna Bruce
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Véronique Picard
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
- Laboratoire d’Hématologie, Faculté de Pharmacie, Université Paris Saclay, Amiens, France
| | - Loïc Garçon
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
- INSERM U1134, INTS, Paris, France
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
16
|
Liu W, Wen D, Liu Z, Wang K, Wang J. Erythropoiesis signature and ubiquitin‐mediated proteolysis are enriched in systematic juvenile idiopathic arthritis. Int J Immunogenet 2022; 49:193-201. [PMID: 35253998 DOI: 10.1111/iji.12573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 02/10/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Wenping Liu
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Dawei Wen
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Ziyi Liu
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Kunyu Wang
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Jibo Wang
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
17
|
Susanah S, Widowati W, Sari NM, Revika R, Kusuma H, Rizal R, Faried A. Potential Use of Patient-Specific Induced Pluripotent Stem Cell for Liver Fibrosis Thalassemia Treatment Management. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Thalassemia is the most common inherited single gene blood disease worldwide and present a significant health problem in the world. Approximately, 1.5% of the global populations (An estimated 80–90 million people) are carriers of β-thalassemia. Around 5% of Indonesia population is thought to carry the thalassemia gene. The globin imbalance in β-thalassemia major causes hemolysis and ineffective erythropoiesis which results in anemia leading to increases of iron absorption. Furthermore, repeated blood transfusion and long-term increased iron absorption will lead to excessive accumulation of iron in vital organs, especially in the liver, causes liver fibrosis then leading to liver disease. Iron overload can be controlled by iron chelating drugs with the risk of side effects; therefore, a breakthrough is needed. Stem cell technology has a potential to provide novel insight in thalassemia major, through induced pluripotent stem cells (iPSCs) who has the ability to differentiate into hepatic stellate cells (HSCs)-like cells. iPSCs derived HSC-like cells (iPSC-HSCs) present the phenotypic and functional characteristics of HSCs. The utilization of iPSCs is a new option in personalized thalassemia management.
Collapse
|
18
|
Tupta B, Stuehr E, Sumi MP, Sweeny EA, Smith B, Stuehr DJ, Ghosh A. GAPDH is involved in the heme-maturation of myoglobin and hemoglobin. FASEB J 2022; 36:e22099. [PMID: 34972240 DOI: 10.1096/fj.202101237rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022]
Abstract
GAPDH, a heme chaperone, has been previously implicated in the incorporation of heme into iNOS and soluble guanylyl cyclase (sGC). Since sGC is critical for myoglobin (Mb) heme-maturation, we investigated the role of GAPDH in the maturation of this globin, as well as hemoglobins α, β, and γ. Utilizing cell culture systems, we found that overexpression of wild-type GAPDH increased, whereas GAPDH mutants H53A and K227A decreased, the heme content of Mb and Hbα and Hbβ. Overexpression of wild-type GAPDH fully recovered the heme-maturation inhibition observed with the GAPDH mutants. Partial rescue was observed by overexpression of sGCβ1 but not by overexpression of a sGCΔβ1 deletion mutant, which is unable to bind the sGCα1 subunit required to form the active sGCα1β1 complex. Wild type and mutant GAPDH was found to be associated in a complex with each of the globins and Hsp90. GAPDH at endogenous levels was found to be associated with Mb in differentiating C2C12 myoblasts, and with Hbγ or Hbα in differentiating HiDEP-1 erythroid progenitor cells. Knockdown of GAPDH in C2C12 cells suppressed Mb heme-maturation. GAPDH knockdown in K562 erythroleukemia cells suppressed Hbα and Hbγ heme-maturation as well as Hb dimerization. Globin heme incorporation was not only dependent on elevated sGCα1β1 heterodimer formation, but also influenced by iron provision and magnitude of expression of GAPDH, d-aminolevulinic acid, and FLVCR1b. Together, our data support an important role for GAPDH in the maturation of myoglobin and γ, β, and α hemoglobins.
Collapse
Affiliation(s)
- Blair Tupta
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Eric Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Mamta P Sumi
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Elizabeth A Sweeny
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Brandon Smith
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
19
|
Fujii J, Homma T, Kobayashi S, Warang P, Madkaikar M, Mukherjee MB. Erythrocytes as a preferential target of oxidative stress in blood. Free Radic Res 2021; 55:562-580. [PMID: 33427524 DOI: 10.1080/10715762.2021.1873318] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Red blood cells (RBC) are specifically differentiated to transport oxygen and carbon dioxide in the blood and they lack most organelles, including mitochondria. The autoxidation of hemoglobin constitutes a major source of reactive oxygen species (ROS). Nitric oxide, which is produced by endothelial nitric oxide synthase (NOS3) or via the hemoglobin-mediated conversion of nitrite, interacts with ROS and results in the production of reactive nitrogen oxide species. Herein we present an overview of anemic diseases that are closely related to oxidative damage. Because the compensation of proteins by means of gene expression does not proceed in enucleated cells, antioxidative and redox systems play more important roles in maintaining the homeostasis of RBC against oxidative insult compared to ordinary cells. Defects in hemoglobin and enzymes that are involved in energy production and redox reactions largely trigger oxidative damage to RBC. The results of studies using genetically modified mice suggest that antioxidative enzymes, notably superoxide dismutase 1 and peroxiredoxin 2, play essential roles in coping with oxidative damage in erythroid cells, and their absence limits erythropoiesis, the life-span of RBC and consequently results in the development of anemia. The degeneration of the machinery involved in the proteolytic removal of damaged proteins appears to be associated with hemolytic events. The ubiquitin-proteasome system is the dominant machinery, not only for the proteolytic removal of damaged proteins in erythroid cells but also for the development of erythropoiesis. Hence, despite the fact that it is less abundant in RBC compared to ordinary cells, the aberrant ubiquitin-proteasome system may be associated with the development of anemic diseases via the accumulation of damaged proteins, as typified in sickle cell disease, and impaired erythropoiesis.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Sho Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Prashant Warang
- ICMR - National Institute of Immunohaematology, Mumbai, India
| | | | | |
Collapse
|
20
|
Lechauve C, Keith J, Khandros E, Fowler S, Mayberry K, Freiwan A, Thom CS, Delbini P, Romero EB, Zhang J, Motta I, Tillman H, Cappellini MD, Kundu M, Weiss MJ. The autophagy-activating kinase ULK1 mediates clearance of free α-globin in β-thalassemia. Sci Transl Med 2020; 11:11/506/eaav4881. [PMID: 31434755 DOI: 10.1126/scitranslmed.aav4881] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/26/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
In β-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate β-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 β-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 β-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of β-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating β-thalassemia.
Collapse
Affiliation(s)
- Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Julia Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Eugene Khandros
- Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie Fowler
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kalin Mayberry
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abdullah Freiwan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher S Thom
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paola Delbini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Emilio Boada Romero
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jingjing Zhang
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Irene Motta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Heather Tillman
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Mondira Kundu
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
21
|
Martinez PA, Li R, Ramanathan HN, Bhasin M, Pearsall RS, Kumar R, Suragani RNVS. Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine β-thalassaemia by increasing GATA-1 availability. J Cell Mol Med 2020; 24:6162-6177. [PMID: 32351032 PMCID: PMC7294138 DOI: 10.1111/jcmm.15243] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 12/16/2022] Open
Abstract
In β‐thalassaemia, anaemia results from ineffective erythropoiesis characterized by inhibition of late‐stage erythroid differentiation. We earlier used luspatercept and RAP‐536 protein traps for certain Smad2/3‐pathway ligands to implicate Smad2/3‐pathway overactivation in dysregulated erythroid differentiation associated with murine β‐thalassaemia and myelodysplasia. Importantly, luspatercept alleviates anaemia and has been shown to reduce transfusion burden in patients with β‐thalassaemia or myelodysplasia. Here, we investigated the molecular mechanisms underlying luspatercept action and pSmad2/3‐mediated inhibition of erythroid differentiation. In murine erythroleukemic (MEL) cells in vitro, ligand‐mediated overactivation of the Smad2/3 pathway reduced nuclear levels of GATA‐1 (GATA‐binding factor‐1) and its transcriptional activator TIF1γ (transcription intermediary factor 1γ), increased levels of reactive oxygen species, reduced cell viability and haemoglobin levels, and inhibited erythroid differentiation. Co‐treatment with luspatercept in MEL cells partially or completely restored each of these. In β‐thalassaemic mice, RAP‐536 up‐regulated Gata1 and its target gene signature in erythroid precursors determined by transcriptional profiling and gene set enrichment analysis, restored nuclear levels of GATA‐1 in erythroid precursors, and nuclear distribution of TIF1γ in erythroblasts. Bone marrow cells from β‐thalassaemic mice treated with luspatercept also exhibited restored nuclear availability of GATA‐1 ex vivo. Our results implicate GATA‐1, and likely TIF1γ, as key mediators of luspatercept/RAP‐536 action in alleviating ineffective erythropoiesis.
Collapse
Affiliation(s)
| | - Robert Li
- Acceleron Pharma, Cambridge, MA, USA
| | | | - Manoj Bhasin
- BIDMC Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, MA, USA
| | | | | | | |
Collapse
|
22
|
Gutiérrez L, Caballero N, Fernández-Calleja L, Karkoulia E, Strouboulis J. Regulation of GATA1 levels in erythropoiesis. IUBMB Life 2019; 72:89-105. [PMID: 31769197 DOI: 10.1002/iub.2192] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
GATA1 is considered as the "master" transcription factor in erythropoiesis. It regulates at the transcriptional level all aspects of erythroid maturation and function, as revealed by gene knockout studies in mice and by genome-wide occupancies in erythroid cells. The GATA1 protein contains two zinc finger domains and an N-terminal transactivation domain. GATA1 translation results in the production of the full-length protein and of a shorter variant (GATA1s) lacking the N-terminal transactivation domain, which is functionally deficient in supporting erythropoiesis. GATA1 protein abundance is highly regulated in erythroid cells at different levels, including transcription, mRNA translation, posttranslational modifications, and protein degradation, in a differentiation-stage-specific manner. Maintaining high GATA1 protein levels is essential in the early stages of erythroid maturation, whereas downregulating GATA1 protein levels is a necessary step in terminal erythroid differentiation. The importance of maintaining proper GATA1 protein homeostasis in erythropoiesis is demonstrated by the fact that both GATA1 loss and its overexpression result in lethal anemia. Importantly, alterations in any of those GATA1 regulatory checkpoints have been recognized as an important cause of hematological disorders such as dyserythropoiesis (with or without thrombocytopenia), β-thalassemia, Diamond-Blackfan anemia, myelodysplasia, or leukemia. In this review, we provide an overview of the multilevel regulation of GATA1 protein homeostasis in erythropoiesis and of its deregulation in hematological disease.
Collapse
Affiliation(s)
- Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Department of Medicine, Universidad de Oviedo, Oviedo, Spain
| | - Noemí Caballero
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Luis Fernández-Calleja
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Elena Karkoulia
- Institute of Molecular Biology and Biotechnology, Foundation of Research & Technology Hellas, Heraklion, Crete, Greece
| | - John Strouboulis
- Cancer Comprehensive Center, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
23
|
Stagg DB, Whittlesey RL, Li X, Lozovatsky L, Gardenghi S, Rivella S, Finberg KE. Genetic loss of Tmprss6 alters terminal erythroid differentiation in a mouse model of β-thalassemia intermedia. Haematologica 2019; 104:e442-e446. [PMID: 30819909 DOI: 10.3324/haematol.2018.213371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- David B Stagg
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Rebecca L Whittlesey
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiuqi Li
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Larisa Lozovatsky
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sara Gardenghi
- Department of Pediatrics, Division of Hematology-Oncology, Weill Cornell Medical College, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology-Oncology, Weill Cornell Medical College, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA.,Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karin E Finberg
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA .,Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Lechauve C, Butcher JT, Freiwan A, Biwer LA, Keith JM, Good ME, Ackerman H, Tillman HS, Kiger L, Isakson BE, Weiss MJ. Endothelial cell α-globin and its molecular chaperone α-hemoglobin-stabilizing protein regulate arteriolar contractility. J Clin Invest 2018; 128:5073-5082. [PMID: 30295646 DOI: 10.1172/jci99933] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Arteriolar endothelial cell-expressed (EC-expressed) α-globin binds endothelial NOS (eNOS) and degrades its enzymatic product, NO, via dioxygenation, thereby lessening the vasodilatory effects of NO on nearby vascular smooth muscle. Although this reaction potentially affects vascular physiology, the mechanisms that regulate α-globin expression and dioxygenase activity in ECs are unknown. Without β-globin, α-globin is unstable and cytotoxic, particularly in its oxidized form, which is generated by dioxygenation and recycled via endogenous reductases. We show that the molecular chaperone α-hemoglobin-stabilizing protein (AHSP) promotes arteriolar α-globin expression in vivo and facilitates its reduction by eNOS. In Ahsp-/- mice, EC α-globin was decreased by 70%. Ahsp-/- and Hba1-/- mice exhibited similar evidence of increased vascular NO signaling, including arteriolar dilation, blunted α1-adrenergic vasoconstriction, and reduced blood pressure. Purified α-globin bound eNOS or AHSP, but not both together. In ECs in culture, eNOS or AHSP enhanced α-globin expression posttranscriptionally. However, only AHSP prevented oxidized α-globin precipitation in solution. Finally, eNOS reduced AHSP-bound α-globin approximately 6-fold faster than did the major erythrocyte hemoglobin reductases (cytochrome B5 reductase plus cytochrome B5). Our data support a model whereby redox-sensitive shuttling of EC α-globin between AHSP and eNOS regulates EC NO degradation and vascular tone.
Collapse
Affiliation(s)
- Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Joshua T Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Abdullah Freiwan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lauren A Biwer
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Julia M Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Heather S Tillman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Meledin R, Mali SM, Kleifeld O, Brik A. Activity-Based Probes Developed by Applying a Sequential Dehydroalanine Formation Strategy to Expressed Proteins Reveal a Potential α-Globin-Modulating Deubiquitinase. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201800032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Roman Meledin
- Schulich Faculty of Chemistry; Technion Israel Institute of Technology; Haifa 3200008 Israel
| | - Sachitanand M. Mali
- Schulich Faculty of Chemistry; Technion Israel Institute of Technology; Haifa 3200008 Israel
| | - Oded Kleifeld
- Faculty of Biology; Technion Israel Institute of Technology; Haifa 3200003 Israel
| | - Ashraf Brik
- Schulich Faculty of Chemistry; Technion Israel Institute of Technology; Haifa 3200008 Israel
| |
Collapse
|
26
|
Meledin R, Mali SM, Kleifeld O, Brik A. Activity-Based Probes Developed by Applying a Sequential Dehydroalanine Formation Strategy to Expressed Proteins Reveal a Potential α-Globin-Modulating Deubiquitinase. Angew Chem Int Ed Engl 2018. [PMID: 29527788 DOI: 10.1002/anie.201800032] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We report a general and novel semisynthetic strategy for the preparation of ubiquitinated protein-activity-based probes on the basis of sequential dehydroalanine formation on expressed proteins. We applied this approach to construct a physiologically and therapeutically relevant ubiquitinated α-globin probe, which was used for the enrichment and proteomic identification of α-globin-modulating deubiquitinases. We found USP15 as a potential deubiquitinase for the modulation of α-globin, an excess of which aggravates β-thalassemia symptoms. This development opens new opportunities for activity-based-probe design to shed light on the important aspects underlying ubiquitination and deubiquitination in health and disease.
Collapse
Affiliation(s)
- Roman Meledin
- Schulich Faculty of Chemistry, Technion Israel Institute of Technology, Haifa, 3200008, Israel
| | - Sachitanand M Mali
- Schulich Faculty of Chemistry, Technion Israel Institute of Technology, Haifa, 3200008, Israel
| | - Oded Kleifeld
- Faculty of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion Israel Institute of Technology, Haifa, 3200008, Israel
| |
Collapse
|
27
|
Hsp90 chaperones hemoglobin maturation in erythroid and nonerythroid cells. Proc Natl Acad Sci U S A 2018; 115:E1117-E1126. [PMID: 29358373 DOI: 10.1073/pnas.1717993115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Maturation of adult (α2β2) and fetal hemoglobin (α2γ2) tetramers requires that heme be incorporated into each globin. While hemoglobin alpha (Hb-α) relies on a specific erythroid chaperone (alpha Hb-stabilizing protein, AHSP), the other chaperones that may help mature the partner globins (Hb-γ or Hb-β) in erythroid cells, or may enable nonerythroid cells to express mature Hb, are unknown. We investigated a role for heat-shock protein 90 (hsp90) in Hb maturation in erythroid precursor cells that naturally express Hb-α with either Hb-γ (K562 and HiDEP-1 cells) or Hb-β (HUDEP-2) and in nonerythroid cell lines that either endogenously express Hb-αβ (RAW and A549) or that we transfected to express the globins. We found the following: (i) AHSP and hsp90 associate with distinct globin partners in their immature heme-free states (AHSP with apo-Hbα, and hsp90 with apo-Hbβ or Hb-γ) and that hsp90 does not associate with mature Hb. (ii) Hsp90 stabilizes the apo-globins and helps to drive their heme insertion reactions, as judged by pharmacologic hsp90 inhibition or by coexpression of an ATP-ase defective hsp90. (iii) In nonerythroid cells, heme insertion into all globins became hsp90-dependent, which may explain how mixed Hb tetramers can mature in cells that do not express AHSP. Together, our findings uncover a process in which hsp90 first binds to immature, heme-free Hb-γ or Hb-β, drives their heme insertion process, and then dissociates to allow their heterotetramer formation with Hb-α. Thus, in driving heme insertion, hsp90 works in concert with AHSP to generate functional Hb tetramers during erythropoiesis.
Collapse
|
28
|
Thanuthanakhun N, Nuntakarn L, Sampattavanich S, Anurathapan U, Phuphanitcharoenkun S, Pornpaiboonstid S, Borwornpinyo S, Hongeng S. Investigation of FoxO3 dynamics during erythroblast development in β-thalassemia major. PLoS One 2017; 12:e0187610. [PMID: 29099866 PMCID: PMC5669432 DOI: 10.1371/journal.pone.0187610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/23/2017] [Indexed: 02/08/2023] Open
Abstract
The FoxO3 transcription factor is a key regulator of oxidative stress and erythroid maturation during erythropoiesis. In this study, we explored the involvement of FoxO3 in severe β-thalassemia. Using primary CD34+ hematopoietic progenitor cells from patients with β-thalassemia major, we successfully developed an in vitro model of ineffective erythropoiesis. Based on this model, FoxO3 activity was quantified in single cells using high throughput imaging flow cytometry. This study revealed a significant reduction of FoxO3 activity during the late stage of erythroblast differentiation in β-thalassemia, in contrast to erythropoiesis in normal cells that maintain persistent activation of FoxO3. In agreement with the decreased FoxO3 activity in β-thalassemia, the expression of FoxO3 target genes was also found to decrease, concurrent with elevated phosphorylation of AKT, most clearly at the late stage of erythroid differentiation. Our findings provide further evidence for the involvement of FoxO3 during terminal erythropoiesis and confirm the modulation of the PI3K/AKT pathway as a potential therapeutic strategy for β-thalassemia.
Collapse
Affiliation(s)
| | - Lalana Nuntakarn
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Somponnat Sampattavanich
- Siriraj Laboratory for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | | | - Suparerk Borwornpinyo
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail: (SB); (SH)
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail: (SB); (SH)
| |
Collapse
|
29
|
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: 1.0] [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
|
30
|
Hodonsky CJ, Jain D, Schick UM, Morrison JV, Brown L, McHugh CP, Schurmann C, Chen DD, Liu YM, Auer PL, Laurie CA, Taylor KD, Browning BL, Li Y, Papanicolaou G, Rotter JI, Kurita R, Nakamura Y, Browning SR, Loos RJF, North KE, Laurie CC, Thornton TA, Pankratz N, Bauer DE, Sofer T, Reiner AP. Genome-wide association study of red blood cell traits in Hispanics/Latinos: The Hispanic Community Health Study/Study of Latinos. PLoS Genet 2017; 13:e1006760. [PMID: 28453575 PMCID: PMC5428979 DOI: 10.1371/journal.pgen.1006760] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 05/12/2017] [Accepted: 04/12/2017] [Indexed: 01/13/2023] Open
Abstract
Prior GWAS have identified loci associated with red blood cell (RBC) traits in populations of European, African, and Asian ancestry. These studies have not included individuals with an Amerindian ancestral background, such as Hispanics/Latinos, nor evaluated the full spectrum of genomic variation beyond single nucleotide variants. Using a custom genotyping array enriched for Amerindian ancestral content and 1000 Genomes imputation, we performed GWAS in 12,502 participants of Hispanic Community Health Study and Study of Latinos (HCHS/SOL) for hematocrit, hemoglobin, RBC count, RBC distribution width (RDW), and RBC indices. Approximately 60% of previously reported RBC trait loci generalized to HCHS/SOL Hispanics/Latinos, including African ancestral alpha- and beta-globin gene variants. In addition to the known 3.8kb alpha-globin copy number variant, we identified an Amerindian ancestral association in an alpha-globin regulatory region on chromosome 16p13.3 for mean corpuscular volume and mean corpuscular hemoglobin. We also discovered and replicated three genome-wide significant variants in previously unreported loci for RDW (SLC12A2 rs17764730, PSMB5 rs941718), and hematocrit (PROX1 rs3754140). Among the proxy variants at the SLC12A2 locus we identified rs3812049, located in a bi-directional promoter between SLC12A2 (which encodes a red cell membrane ion-transport protein) and an upstream anti-sense long-noncoding RNA, LINC01184, as the likely causal variant. We further demonstrate that disruption of the regulatory element harboring rs3812049 affects transcription of SLC12A2 and LINC01184 in human erythroid progenitor cells. Together, these results reinforce the importance of genetic study of diverse ancestral populations, in particular Hispanics/Latinos.
Collapse
Affiliation(s)
- Chani J. Hodonsky
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, NC, United States of America
| | - Deepti Jain
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Ursula M. Schick
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jean V. Morrison
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Lisa Brown
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Caitlin P. McHugh
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
- New York Genome Center, New York, NY, United States of America
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Diane D. Chen
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States of America
| | - Yong Mei Liu
- School of Medicine, Wake Forest University, Winston-Salem, NC, United States of America
| | - Paul L. Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin Milwaukee, Milwaukee, WI, United States of America
| | - Cecilia A. Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA United States of America
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA United States of America
| | - Brian L. Browning
- Department of Medicine, University of Washington, Seattle, WA United States of America
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, United States of America
| | - George Papanicolaou
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA United States of America
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA United States of America
| | - Ryo Kurita
- Research and Development Department, Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
- Comprehensive Human Sciences, Tsukuba University, Tsukuba, Ibaraki, Japan
| | - Sharon R. Browning
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kari E. North
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, NC, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America
| | - Cathy C. Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Nathan Pankratz
- Division of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Daniel E. Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States of America
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
- Department of Pediatrics, Harvard Medical School and Harvard Stem Cell Institute, Harvard University, Boston, MA, United States of America
| | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW The review provides an overview of recent data regarding the molecular players in β-thalassemia dyserythropoiesis and the corresponding therapeutic implications. RECENT FINDINGS β-thalassemia dyserythropoiesis is characterized by four steps: expansion of erythroid progenitors, accelerated erythroid differentiation until the polychromatophilic stage, maturation arrest, and apoptosis at the polychromatophilic stage. Excess α-globin chains are the primary culprit in the disease, but the link between this excess and ineffective erythropoiesis has only recently been established. Important recent advances in understanding the molecular determinants involved in two critical steps of dyserythropoiesis are paving the way to new alternative targets for the treatment of this disease. SUMMARY Growth differentiation factor 11 (GDF11) blockade increases the apoptosis of erythroblasts with excess α-chains by upregulating Fas-ligand in late basophilic and polychromatophilic erythroblasts, thereby decreasing cell expansion (step 1). Blocking GDF11 alleviates anemia in a mouse model of β-thalassemia and also in humans, most likely by promoting cells of 'good' erythroblastic lineage containing an α-/non-α-globin chain ratio of close to 1. Maturation arrest at the polychromatophilic stage (step 3) is associated with the depletion of GATA binding protein 1 (GATA-1) from the nucleus, which results from cytoplasmic sequestration of heat shock protein 70 (HSP70) by α-globin chains. Small molecules disrupting the HSP70/α-globin complex in the cytoplasm or decreasing HSP70 nuclear export might increase the nuclear localization of HSP70, thereby protecting GATA-1 and alleviating anemia. Finally, increasing the serum levels of hepcidin or transferrin alleviates anemia and dyserythropoiesis by diminishing iron uptake by erythroblasts in mouse models.
Collapse
|
32
|
miR-150 inhibits terminal erythroid proliferation and differentiation. Oncotarget 2016; 6:43033-47. [PMID: 26543232 PMCID: PMC4767489 DOI: 10.18632/oncotarget.5824] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/22/2015] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs), a class of small non-coding linear RNAs, have been shown to play a crucial role in erythropoiesis. To evaluate the indispensable role of constant suppression of miR-150 during terminal erythropoiesis, we performed miR-150 gain- and loss-of-function experiments on hemin-induced K562 cells and EPO-induced human CD34+ cells. We found that forced expression of miR-150 suppresses commitment of hemoglobinization and CD235a labeling in both cell types. Erythroid proliferation is also inhibited via inducing apoptosis and blocking the cell cycle when miR-150 is overexpressed. In contrast, miR-150 inhibition promotes terminal erythropoiesis. 4.1 R gene is a new target of miR-150 during terminal erythropoiesis, and its abundance ensures the mechanical stability and deformability of the membrane. However, knockdown of 4.1 R did not affect terminal erythropoiesis. Transcriptional profiling identified more molecules involved in terminal erythroid dysregulation derived from miR-150 overexpression. These results shed light on the role of miR-150 during human terminal erythropoiesis. This is the first report highlighting the relationship between miRNA and membrane protein and enhancing our understanding of how miRNA works in the hematopoietic system.
Collapse
|
33
|
Pomalidomide reverses γ-globin silencing through the transcriptional reprogramming of adult hematopoietic progenitors. Blood 2015; 127:1481-92. [PMID: 26679864 DOI: 10.1182/blood-2015-09-667923] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/02/2015] [Indexed: 12/24/2022] Open
Abstract
Current therapeutic strategies for sickle cell anemia are aimed at reactivating fetal hemoglobin. Pomalidomide, a third-generation immunomodulatory drug, was proposed to induce fetal hemoglobin production by an unknown mechanism. Here, we report that pomalidomide induced a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. Pomalidomide acted early by transiently delaying erythropoiesis at the burst-forming unit-erythroid/colony-forming unit-erythroid transition, but without affecting terminal differentiation. Further, the transcription networks involved in γ-globin repression were selectively and differentially affected by pomalidomide including BCL11A, SOX6, IKZF1, KLF1, and LSD1. IKAROS (IKZF1), a known target of pomalidomide, was degraded by the proteasome, but was not the key effector of this program, because genetic ablation of IKZF1 did not phenocopy pomalidomide treatment. Notably, the pomalidomide-induced reprogramming was conserved in hematopoietic progenitors from individuals with sickle cell anemia. Moreover, multiple myeloma patients treated with pomalidomide demonstrated increased in vivo γ-globin levels in their erythrocytes. Together, these data reveal the molecular mechanisms by which pomalidomide reactivates fetal hemoglobin, reinforcing its potential as a treatment for patients with β-hemoglobinopathies.
Collapse
|
34
|
Voskou S, Aslan M, Fanis P, Phylactides M, Kleanthous M. Oxidative stress in β-thalassaemia and sickle cell disease. Redox Biol 2015; 6:226-239. [PMID: 26285072 PMCID: PMC4543215 DOI: 10.1016/j.redox.2015.07.018] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022] Open
Abstract
Sickle cell disease and β-thalassaemia are inherited haemoglobinopathies resulting in structural and quantitative changes in the β-globin chain. These changes lead to instability of the generated haemoglobin or to globin chain imbalance, which in turn impact the oxidative environment both intracellularly and extracellularly. The ensuing oxidative stress and the inability of the body to adequately overcome it are, to a large extent, responsible for the pathophysiology of these diseases. This article provides an overview of the main players and control mechanisms involved in the establishment of oxidative stress in these haemoglobinopathies.
Collapse
Affiliation(s)
- S Voskou
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - M Aslan
- Akdeniz University, Faculty of Medicine, Department of Medical Biochemistry, Antalya, Turkey
| | - P Fanis
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - M Phylactides
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.
| | - M Kleanthous
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
35
|
Thom CS, Traxler EA, Khandros E, Nickas JM, Zhou OY, Lazarus JE, Silva APG, Prabhu D, Yao Y, Aribeana C, Fuchs SY, Mackay JP, Holzbaur ELF, Weiss MJ. Trim58 degrades Dynein and regulates terminal erythropoiesis. Dev Cell 2014; 30:688-700. [PMID: 25241935 DOI: 10.1016/j.devcel.2014.07.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 04/24/2014] [Accepted: 07/28/2014] [Indexed: 01/23/2023]
Abstract
TRIM58 is an E3 ubiquitin ligase superfamily member implicated by genome-wide association studies to regulate human erythrocyte traits. Here, we show that Trim58 expression is induced during late erythropoiesis and that its depletion by small hairpin RNAs (shRNAs) inhibits the maturation of late-stage nucleated erythroblasts to anucleate reticulocytes. Imaging flow cytometry studies demonstrate that Trim58 regulates polarization and/or extrusion of erythroblast nuclei. In vitro, Trim58 directly binds and ubiquitinates the intermediate chain of the microtubule motor dynein. In cells, Trim58 stimulates proteasome-dependent degradation of the dynein holoprotein complex. During erythropoiesis, Trim58 expression, dynein loss, and enucleation occur concomitantly, and all are inhibited by Trim58 shRNAs. Dynein regulates nuclear positioning and microtubule organization, both of which undergo dramatic changes during erythroblast enucleation. Thus, we propose that Trim58 promotes this process by eliminating dynein. Our findings identify an erythroid-specific regulator of enucleation and elucidate a previously unrecognized mechanism for controlling dynein activity.
Collapse
Affiliation(s)
- Christopher S Thom
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth A Traxler
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eugene Khandros
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jenna M Nickas
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Olivia Y Zhou
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jacob E Lazarus
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana P G Silva
- School of Molecular Bioscience, The University of Sydney, Sydney NSW 2006, Australia
| | - Dolly Prabhu
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yu Yao
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chiaka Aribeana
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Animal Biology and Mari Lowe Comparative Oncology Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joel P Mackay
- School of Molecular Bioscience, The University of Sydney, Sydney NSW 2006, Australia
| | - Erika L F Holzbaur
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell J Weiss
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Arlet JB, Ribeil JA, Guillem F, Negre O, Hazoume A, Marcion G, Beuzard Y, Dussiot M, Moura IC, Demarest S, de Beauchêne IC, Belaid-Choucair Z, Sevin M, Maciel TT, Auclair C, Leboulch P, Chretien S, Tchertanov L, Baudin-Creuza V, Seigneuric R, Fontenay M, Garrido C, Hermine O, Courtois G. HSP70 sequestration by free α-globin promotes ineffective erythropoiesis in β-thalassaemia. Nature 2014; 514:242-6. [PMID: 25156257 DOI: 10.1038/nature13614] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/25/2014] [Indexed: 12/28/2022]
Abstract
β-Thalassaemia major (β-TM) is an inherited haemoglobinopathy caused by a quantitative defect in the synthesis of β-globin chains of haemoglobin, leading to the accumulation of free α-globin chains that form toxic aggregates. Despite extensive knowledge of the molecular defects causing β-TM, little is known of the mechanisms responsible for the ineffective erythropoiesis observed in the condition, which is characterized by accelerated erythroid differentiation, maturation arrest and apoptosis at the polychromatophilic stage. We have previously demonstrated that normal human erythroid maturation requires a transient activation of caspase-3 at the later stages of maturation. Although erythroid transcription factor GATA-1, the master transcriptional factor of erythropoiesis, is a caspase-3 target, it is not cleaved during erythroid differentiation. We have shown that, in human erythroblasts, the chaperone heat shock protein70 (HSP70) is constitutively expressed and, at later stages of maturation, translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. The primary role of this ubiquitous chaperone is to participate in the refolding of proteins denatured by cytoplasmic stress, thus preventing their aggregation. Here we show in vitro that during the maturation of human β-TM erythroblasts, HSP70 interacts directly with free α-globin chains. As a consequence, HSP70 is sequestrated in the cytoplasm and GATA-1 is no longer protected, resulting in end-stage maturation arrest and apoptosis. Transduction of a nuclear-targeted HSP70 mutant or a caspase-3-uncleavable GATA-1 mutant restores terminal maturation of β-TM erythroblasts, which may provide a rationale for new targeted therapies of β-TM.
Collapse
Affiliation(s)
- Jean-Benoît Arlet
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Service de Médecine Interne, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 15 rue Leblanc 75908 Paris, France [3] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [4] Laboratory of Excellence GR-Ex, 75015 Paris, France [5]
| | - Jean-Antoine Ribeil
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] Département de Biothérapie, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris, Hôpital Necker, 149 rue de Sèvres 75015 Paris, France [5]
| | - Flavia Guillem
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France
| | - Olivier Negre
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Adonis Hazoume
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Guillaume Marcion
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Yves Beuzard
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Michaël Dussiot
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France
| | - Ivan Cruz Moura
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France [5] Faculté de médecine and Université Denis Diderot Paris VII, 5 Rue Thomas Mann, 75013 Paris, France
| | - Samuel Demarest
- Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France
| | - Isaure Chauvot de Beauchêne
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | - Zakia Belaid-Choucair
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France
| | - Margaux Sevin
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Thiago Trovati Maciel
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France [5] Faculté de médecine and Université Denis Diderot Paris VII, 5 Rue Thomas Mann, 75013 Paris, France
| | - Christian Auclair
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | - Philippe Leboulch
- 1] Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France [2] Women's Hospital and Harvard Medical School, 25 Shattuck St, Boston, Massachusetts 02115, USA
| | - Stany Chretien
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Luba Tchertanov
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | | | - Renaud Seigneuric
- University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Michaela Fontenay
- 1] Laboratory of Excellence GR-Ex, 75015 Paris, France [2] Institut Cochin, INSERM, unité mixte de recherche 1016, centre national de la recherche scientifique (CNRS), unité mixte de recherche 8104, Université Paris Descartes, and Assistance publique - Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Service d'hématologie biologique, 27 rue du Faubourg Saitn-Jacques, 75014 Paris, France
| | - Carmen Garrido
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France [3] Centre anticancéreux George François Leclerc, 1 rue professeur Marion, 21079 Dijon, France [4]
| | - Olivier Hermine
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] Service d'hématologie, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris Hôpital Necker, 149 rue de Sèvres, 75015 Paris, France [5]
| | - Geneviève Courtois
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4]
| |
Collapse
|
37
|
Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine β-thalassemia. Blood 2014; 123:3864-72. [PMID: 24795345 DOI: 10.1182/blood-2013-06-511238] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In β-thalassemia, unequal production of α- and β-globin chains in erythroid precursors causes apoptosis and inhibition of late-stage erythroid differentiation, leading to anemia, ineffective erythropoiesis (IE), and dysregulated iron homeostasis. Here we used a murine model of β-thalassemia intermedia (Hbb(th1/th1) mice) to investigate effects of a modified activin receptor type IIB (ActRIIB) ligand trap (RAP-536) that inhibits Smad2/3 signaling. In Hbb(th1/th1) mice, treatment with RAP-536 reduced overactivation of Smad2/3 in splenic erythroid precursors. In addition, treatment of Hbb(th1/th1) mice with RAP-536 reduced α-globin aggregates in peripheral red cells, decreased the elevated reactive oxygen species present in erythroid precursors and peripheral red cells, and alleviated anemia by promoting differentiation of late-stage erythroid precursors and reducing hemolysis. Notably, RAP-536 treatment mitigated disease complications of IE, including iron overload, splenomegaly, and bone pathology, while reducing erythropoietin levels, improving erythrocyte morphology, and extending erythrocyte life span. These results implicate signaling by the transforming growth factor-β superfamily in late-stage erythropoiesis and reveal potential of a modified ActRIIB ligand trap as a novel therapeutic agent for thalassemia syndrome and other red cell disorders characterized by IE.
Collapse
|
38
|
Kiger L, Vasseur C, Domingues-Hamdi E, Truan G, Marden MC, Baudin-Creuza V. Dynamics of α-Hb chain binding to its chaperone AHSP depends on heme coordination and redox state. Biochim Biophys Acta Gen Subj 2014; 1840:277-87. [DOI: 10.1016/j.bbagen.2013.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 11/27/2022]
|
39
|
Erythropoietic defect associated with reduced cell proliferation in mice lacking the 26S proteasome shuttling factor Rad23b. Mol Cell Biol 2013; 33:3879-92. [PMID: 23897431 DOI: 10.1128/mcb.05772-11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rad23a and Rad23b proteins are linked to nucleotide excision DNA repair (NER) via association with the DNA damage recognition protein xeroderma pigmentosum group C (XPC) are and known to be implicated in protein turnover by the 26S proteasome. Rad23b-null mice are NER proficient, likely due to the redundant function of the Rad23b paralogue, Rad23a. However, Rad23b-null midgestation embryos are anemic, and most embryos die before birth. Using an unbiased proteomics approach, we found that the majority of Rad23b-interacting partners are associated with the ubiquitin-proteasome system (UPS). We tested the requirement for Rad23b-dependent UPS activity in cellular proliferation and more specifically in the process of erythropoiesis. In cultured fibroblasts derived from embryos lacking Rad23b, proliferation rates were reduced. In fetal livers of Rad23b-null embryos, we observed reduced proliferation, accumulation of early erythroid progenitors, and a block during erythroid maturation. In primary wild-type (WT) erythroid cells, knockdown of Rad23b or chemical inhibition of the proteasome reduced survival and differentiation capability. Finally, the defects linked to Rad23b loss specifically affected fetal definitive erythropoiesis and stress erythropoiesis in adult mice. Together, these data indicate a previously unappreciated requirement for Rad23b and the UPS in regulation of proliferation in different cell types.
Collapse
|
40
|
Ineffective erythropoiesis in β -thalassemia. ScientificWorldJournal 2013; 2013:394295. [PMID: 23606813 PMCID: PMC3628659 DOI: 10.1155/2013/394295] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 02/03/2013] [Indexed: 01/06/2023] Open
Abstract
In humans, β-thalassemia dyserythropoiesis is characterized by expansion of early erythroid precursors and erythroid progenitors and then ineffective erythropoiesis. This ineffective erythropoiesis is defined as a suboptimal production of mature erythrocytes originating from a proliferating pool of immature erythroblasts. It is characterized by (1) accelerated erythroid differentiation, (2) maturation blockade at the polychromatophilic stage, and (3) death of erythroid precursors. Despite extensive knowledge of molecular defects causing β-thalassemia, less is known about the mechanisms responsible for ineffective erythropoiesis. In this paper, we will focus on the underlying mechanisms leading to premature death of thalassemic erythroid precursors in the bone marrow.
Collapse
|
41
|
Characterization of the TGF-β1 signaling abnormalities in the Gata1low mouse model of myelofibrosis. Blood 2013; 121:3345-63. [PMID: 23462118 DOI: 10.1182/blood-2012-06-439661] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Primary myelofibrosis (PMF) is characterized by fibrosis, ineffective hematopoiesis in marrow, and hematopoiesis in extramedullary sites and is associated with abnormal megakaryocyte (MK) development and increased transforming growth factor (TGF)-β1 release. To clarify the role of TGF-β1 in the pathogenesis of this disease, the TGF-β1 signaling pathway of marrow and spleen of the Gata1(low) mouse model of myelofibrosis (MF) was profiled and the consequences of inhibition of TGF-β1 signaling on disease manifestations determined. The expression of 20 genes in marrow and 36 genes in spleen of Gata1(low) mice was altered. David-pathway analyses identified alterations of TGF-β1, Hedgehog, and p53 signaling in marrow and spleen and of mammalian target of rapamycin (mTOR) in spleen only and predicted that these alterations would induce consequences consistent with the Gata1(low) phenotype (increased apoptosis and G1 arrest both in marrow and spleen and increased osteoblast differentiation and reduced ubiquitin-mediated proteolysis in marrow only). Inhibition of TGF-β1 signaling normalized the expression of p53-related genes, restoring hematopoiesis and MK development and reducing fibrosis, neovascularization, and osteogenesis in marrow. It also normalized p53/mTOR/Hedgehog-related genes in spleen, reducing extramedullary hematopoiesis. These data identify altered expression signatures of TGF-β1 signaling that may be responsible for MF in Gata1(low) mice and may represent additional targets for therapeutic intervention in PMF.
Collapse
|
42
|
Nienhuis AW, Nathan DG. Pathophysiology and Clinical Manifestations of the β-Thalassemias. Cold Spring Harb Perspect Med 2012; 2:a011726. [PMID: 23209183 DOI: 10.1101/cshperspect.a011726] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The β-thalassemia syndromes reflect deficient or absent β-globin synthesis usually owing to a mutation in the β-globin locus. The relative excess of α-globin results in the formation of insoluble aggregates leading to ineffective erythropoiesis and shortened red cell survival. A relatively high capacity for fetal hemoglobin synthesis is a major genetic modifier of disease severity, with polymorphisms in other genes also having a significant role. Iron overload secondary to enhanced absorption and red cell transfusions causes an increase in liver iron and in various other tissues, leading to endocrine and cardiac dysfunction. Modern chelation regimens are effective in removing iron and preserving or restoring organ function.
Collapse
Affiliation(s)
- Arthur W Nienhuis
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | |
Collapse
|
43
|
Abstract
I have always been puzzled by the ability of normal erythroid cells to produce a stoichiometric amount of α- and β-globin chains in the absence of cross-talk mechanisms that would control and normalize the relative rate of expression of the genes encoding these proteins.
Collapse
|
44
|
Integrated protein quality-control pathways regulate free α-globin in murine β-thalassemia. Blood 2012; 119:5265-75. [PMID: 22427201 DOI: 10.1182/blood-2011-12-397729] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cells remove unstable polypeptides through protein quality-control (PQC) pathways such as ubiquitin-mediated proteolysis and autophagy. In the present study, we investigated how these pathways are used in β-thalassemia, a common hemoglobinopathy in which β-globin gene mutations cause the accumulation and precipitation of cytotoxic α-globin subunits. In β-thalassemic erythrocyte precursors, free α-globin was polyubiquitinated and degraded by the proteasome. These cells exhibited enhanced proteasome activity, and transcriptional profiling revealed coordinated induction of most proteasome subunits that was mediated by the stress-response transcription factor Nrf1. In isolated thalassemic cells, short-term proteasome inhibition blocked the degradation of free α-globin. In contrast, prolonged in vivo treatment of β-thalassemic mice with the proteasome inhibitor bortezomib did not enhance the accumulation of free α-globin. Rather, systemic proteasome inhibition activated compensatory proteotoxic stress-response mechanisms, including autophagy, which cooperated with ubiquitin-mediated proteolysis to degrade free α-globin in erythroid cells. Our findings show that multiple interregulated PQC responses degrade excess α-globin. Therefore, β-thalassemia fits into the broader framework of protein-aggregation disorders that use PQC pathways as cell-protective mechanisms.
Collapse
|
45
|
Aksoy C, Guliyev A, Kilic E, Uckan D, Severcan F. Bone marrow mesenchymal stem cells in patients with beta thalassemia major: molecular analysis with attenuated total reflection-Fourier transform infrared spectroscopy study as a novel method. Stem Cells Dev 2012; 21:2000-11. [PMID: 22214206 DOI: 10.1089/scd.2011.0444] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BM-MSCs) are the main cellular components of the bone marrow, providing a supportive cellular microenvironment to maintain healthy hematopoiesis. β-thalassemia major (β-TM) is characterized by anemia that is caused by a genetic defect in hemoglobin synthesis and results in ineffective erythropoiesis (IE). The alterations in the microenvironment in thalassemic bone marrow during IE can cause changes in BM-MSCs. This study aimed to investigate global structural and compositional changes in BM-MSCs in β-TM that may provide a basis in understanding interactions of hematopoietic stem cells (HSCs)-MSCs in such a pathological bone marrow microenvironment. Following characterization of morphological, immunophenotypical, and differentiation properties, the changes in healthy and thalassemic BM-MSCs before and after bone marrow transplantation (BMT) were examined by attenuated total reflection-Fourier transform infrared (ATR-FTIR). The significant increase in lipid, protein, glycogen, and nucleic acid contents in thalassemic BM-MSCs with respect to healthy BM-MSCs was attributed to enhanced cell proliferation and BM activity during IE. The significant decreases in the content of mentioned macromolecules in post-transplant group BM-MSCs versus pre-transplant BM-MSCs was interpreted as restoring effect of BMT therapy on IE and defective BM microenvironment. These alterations were also supported by ELISA results of erythropoietin (EPO) and growth differentiation factor (GDF15) in bone marrow plasma samples as a reflection of IE and by MTT proliferation assay on BM-MSCs. Based on these changes, sampling groups were discriminated by cluster analysis. These results provide information for the studies that concentrate on interactions between HSCs-MSCs in bone marrow.
Collapse
Affiliation(s)
- Ceren Aksoy
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | | | | | | | | |
Collapse
|
46
|
Khandros E, Mollan TL, Yu X, Wang X, Yao Y, D'Souza J, Gell DA, Olson JS, Weiss MJ. Insights into hemoglobin assembly through in vivo mutagenesis of α-hemoglobin stabilizing protein. J Biol Chem 2012; 287:11325-37. [PMID: 22287545 DOI: 10.1074/jbc.m111.313205] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
α-Hemoglobin stabilizing protein (AHSP) is believed to facilitate adult Hemoglobin A assembly and protect against toxic free α-globin subunits. Recombinant AHSP binds multiple forms of free α-globin to stabilize their structures and inhibit precipitation. However, AHSP also stimulates autooxidation of αO(2) subunit and its rapid conversion to a partially unfolded bishistidyl hemichrome structure. To investigate these biochemical properties, we altered the evolutionarily conserved AHSP proline 30 in recombinantly expressed proteins and introduced identical mutations into the endogenous murine Ahsp gene. In vitro, the P30W AHSP variant bound oxygenated α chains with 30-fold increased affinity. Both P30W and P30A mutant proteins also caused decreased rates of αO(2) autooxidation as compared with wild-type AHSP. Despite these abnormalities, mice harboring P30A or P30W Ahsp mutations exhibited no detectable defects in erythropoiesis at steady state or during induced stresses. Further biochemical studies revealed that the AHSP P30A and P30W substitutions had minimal effects on AHSP interactions with ferric α subunits. Together, our findings indicate that the ability of AHSP to stabilize nascent α chain folding intermediates prior to hemin reduction and incorporation into adult Hemoglobin A is physiologically more important than AHSP interactions with ferrous αO(2) subunits.
Collapse
Affiliation(s)
- Eugene Khandros
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|