1
|
Malay J, Salama RAA, Alam Qureshi GS, Ammar ARAA, Janardhan G, Safdar M, Elshamy HAH. Gene Therapy: A Revolutionary Step in Treating Thalassemia. Hematol Rep 2024; 16:656-668. [PMID: 39449307 PMCID: PMC11503351 DOI: 10.3390/hematolrep16040064] [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: 06/03/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Beta thalassemia is an inherited blood disorder that results in inefficient erythropoiesis due to genetic mutation that leads to the reduction or absence of the hemoglobin beta-globulin protein. Approximately 8.5% of UAE residents suffer from β-thalassemia, a significant health and financial problem. The treatment options available for β-Thalassemia major are limited and associated with a wide range of complications. β-thalassemia gene therapy is emerging as a potential novel treatment option that eliminates the complications caused by the current long-term treatment modalities and the associated economic burden. This paper reviews the scientific literature related to emerging gene therapy for β-Thalassemia by analyzing all the articles published from January 2010 to December 2023 in the English language on Databases like PubMed, Scopus, ProQuest, and CINAHL. The use of gene therapy has demonstrated promising outcomes for a permanent cure of β-Thalassemia. To conclude, gene therapy is an innovative solution. It demonstrates a promising future, but does come with its own setbacks and is something that must be tackled in order to revolutionize it in the medical world. FDA-approved ZYNTEGLO is a potentially one-time curative treatment for β-Thalassemia. Although cutting-edge, its use is limited because of the high cost-a price of USD 2.8 million per patient.
Collapse
Affiliation(s)
- Jhancy Malay
- Department of Pediatrics, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates
| | - Rasha Aziz Attia Salama
- Department of Community Medicine, RAK Medical and Health Science University, Ras Al Khaimah 11127, United Arab Emirates;
| | - Ghania Shehzad Alam Qureshi
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates; (G.S.A.Q.); (A.R.); (G.J.); (M.S.)
| | - Ali Raafat Ali Ahmed Ammar
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates; (G.S.A.Q.); (A.R.); (G.J.); (M.S.)
| | - Gayatri Janardhan
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates; (G.S.A.Q.); (A.R.); (G.J.); (M.S.)
| | - Maryam Safdar
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates; (G.S.A.Q.); (A.R.); (G.J.); (M.S.)
| | - Hesham Amin Hamdy Elshamy
- Department of Surgery, RAK Medical and Health Sciences University, Ras Al Khaimah 11127, United Arab Emirates;
| |
Collapse
|
2
|
Nainggolan I, Amanda N, Batubara I, Saepuloh U, Darusman H. Fetal Hemoglobin Inducer Activity of Moringa oleifera, Curcuma aueruginosa Roxb., and Artocarpus altilis Based on the Gamma Globin Expression. JOURNAL OF APPLIED HEMATOLOGY 2023. [DOI: 10.4103/joah.joah_74_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
|
3
|
Ali S, Mumtaz S, Shakir HA, Khan M, Tahir HM, Mumtaz S, Mughal TA, Hassan A, Kazmi SAR, Sadia, Irfan M, Khan MA. Current status of beta-thalassemia and its treatment strategies. Mol Genet Genomic Med 2021; 9:e1788. [PMID: 34738740 PMCID: PMC8683628 DOI: 10.1002/mgg3.1788] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/15/2020] [Accepted: 08/13/2021] [Indexed: 01/05/2023] Open
Abstract
Background Thalassemia is an inherited hematological disorder categorized by a decrease or absence of one or more of the globin chains synthesis. Beta‐thalassemia is caused by one or more mutations in the beta‐globin gene. The absence or reduced amount of beta‐globin chains causes ineffective erythropoiesis which leads to anemia. Methods Beta‐thalassemia has been further divided into three main forms: thalassemia major, intermedia, and minor/silent carrier. A more severe form among these is thalassemia major in which individuals depend upon blood transfusion for survival. The high level of iron deposition occurs due to regular blood transfusion therapy. Results Overloaded iron raises the synthesis of reactive oxygen species (ROS) that are noxious and prompting the injury to the hepatic, endocrine, and vascular system. Thalassemia can be analyzed and diagnosed via prenatal testing (genetic testing of amniotic fluid), blood smear, complete blood count, and DNA analysis (genetic testing). Treatment of thalassemia intermediate is symptomatic; however; it can also be accomplished by folic supplementation and splenectomy. Conclusion Thalassemia major can be cured through regular transfusion of blood, transplantation of bone marrow, iron chelation management, hematopoietic stem cell transplantation, stimulation of fetal hemoglobin production, and gene therapy.
Collapse
Affiliation(s)
- Shaukat Ali
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shumaila Mumtaz
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Muhammad Khan
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Hafiz Muhammad Tahir
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Samaira Mumtaz
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Tafail Akbar Mughal
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Ali Hassan
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Sadia
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Adeeb Khan
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| |
Collapse
|
4
|
de Melo TRF, Dulmovits BM, Fernandes GFDS, de Souza CM, Lanaro C, He M, Al Abed Y, Chung MC, Blanc L, Costa FF, Dos Santos JL. Synthesis and pharmacological evaluation of pomalidomide derivatives useful for sickle cell disease treatment. Bioorg Chem 2021; 114:105077. [PMID: 34130111 PMCID: PMC8387409 DOI: 10.1016/j.bioorg.2021.105077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Fetal hemoglobin (HbF) induction constitutes a valuable and validated approach to treat the symptoms of sickle cell disease (SCD). Here, we synthesized pomalidomide-nitric oxide (NO) donor derivatives (3a-f) and evaluated their suitability as novel HbF inducers. All compounds demonstrated different capacities of releasing NO, ranging 0.3-30.3%. Compound 3d was the most effective HbF inducer for CD34+ cells, exhibiting an effect similar to that of hydroxyurea. We investigated the mode of action of compound 3d for HbF induction by studying the in vitro alterations in the levels of transcription factors (BCL11A, IKAROS, and LRF), inhibition of histone deacetylase enzymes (HDAC-1 and HDAC-2), and measurement of cGMP levels. Additionally, compound 3d exhibited a potent anti-inflammatory effect similar to that of pomalidomide by reducing the TNF-α levels in human mononuclear cells treated with lipopolysaccharides up to 58.6%. Chemical hydrolysis studies revealed that compound 3d was stable at pH 7.4 up to 24 h. These results suggest that compound 3d is a novel HbF inducer prototype with the potential to treat SCD symptoms.
Collapse
Affiliation(s)
| | - Brian M Dulmovits
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | - Cristiane M de Souza
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Carolina Lanaro
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Minghzu He
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Yousef Al Abed
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Man Chin Chung
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Lionel Blanc
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil.
| |
Collapse
|
5
|
Papasavva PL, Papaioannou NY, Patsali P, Kurita R, Nakamura Y, Sitarou M, Christou S, Kleanthous M, Lederer CW. Distinct miRNA Signatures and Networks Discern Fetal from Adult Erythroid Differentiation and Primary from Immortalized Erythroid Cells. Int J Mol Sci 2021; 22:3626. [PMID: 33807258 PMCID: PMC8037168 DOI: 10.3390/ijms22073626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/22/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs crucial for post-transcriptional and translational regulation of cellular and developmental pathways. The study of miRNAs in erythropoiesis elucidates underlying regulatory mechanisms and facilitates related diagnostic and therapy development. Here, we used DNA Nanoball (DNB) small RNA sequencing to comprehensively characterize miRNAs in human erythroid cell cultures. Based on primary human peripheral-blood-derived CD34+ (hCD34+) cells and two influential erythroid cell lines with adult and fetal hemoglobin expression patterns, HUDEP-2 and HUDEP-1, respectively, our study links differential miRNA expression to erythroid differentiation, cell type, and hemoglobin expression profile. Sequencing results validated by reverse-transcription quantitative PCR (RT-qPCR) of selected miRNAs indicate shared differentiation signatures in primary and immortalized cells, characterized by reduced overall miRNA expression and reciprocal expression increases for individual lineage-specific miRNAs in late-stage erythropoiesis. Despite the high similarity of same-stage hCD34+ and HUDEP-2 cells, differential expression of several miRNAs highlighted informative discrepancies between both cell types. Moreover, a comparison between HUDEP-2 and HUDEP-1 cells displayed changes in miRNAs, transcription factors (TFs), target genes, and pathways associated with globin switching. In resulting TF-miRNA co-regulatory networks, major therapeutically relevant regulators of globin expression were targeted by many co-expressed miRNAs, outlining intricate combinatorial miRNA regulation of globin expression in erythroid cells.
Collapse
Affiliation(s)
- Panayiota L. Papasavva
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.L.P.); (N.Y.P.); (P.P.); (M.K.)
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| | - Nikoletta Y. Papaioannou
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.L.P.); (N.Y.P.); (P.P.); (M.K.)
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| | - Petros Patsali
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.L.P.); (N.Y.P.); (P.P.); (M.K.)
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| | - Ryo Kurita
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan; (R.K.); (Y.N.)
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan; (R.K.); (Y.N.)
| | - Maria Sitarou
- Thalassemia Clinic Larnaca, Larnaca General Hospital, Larnaca 6301, Cyprus;
| | - Soteroulla Christou
- Thalassemia Clinic Nicosia, Archbishop Makarios III Hospital, Nicosia 1474, Cyprus;
| | - Marina Kleanthous
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.L.P.); (N.Y.P.); (P.P.); (M.K.)
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| | - Carsten W. Lederer
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.L.P.); (N.Y.P.); (P.P.); (M.K.)
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| |
Collapse
|
6
|
Breveglieri G, Pacifico S, Zuccato C, Cosenza LC, Sultan S, D’Aversa E, Gambari R, Preti D, Trapella C, Guerrini R, Borgatti M. Discovery of Novel Fetal Hemoglobin Inducers through Small Chemical Library Screening. Int J Mol Sci 2020; 21:E7426. [PMID: 33050052 PMCID: PMC7582302 DOI: 10.3390/ijms21197426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/27/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022] Open
Abstract
The screening of chemical libraries based on cellular biosensors is a useful approach to identify new hits for novel therapeutic targets involved in rare genetic pathologies, such as β-thalassemia and sickle cell disease. In particular, pharmacologically mediated stimulation of human γ-globin gene expression, and increase of fetal hemoglobin (HbF) production, have been suggested as potential therapeutic strategies for these hemoglobinopathies. In this article, we screened a small chemical library, constituted of 150 compounds, using the cellular biosensor K562.GR, carrying enhanced green fluorescence protein (EGFP) and red fluorescence protein (RFP) genes under the control of the human γ-globin and β-globin gene promoters, respectively. Then the identified compounds were analyzed as HbF inducers on primary cell cultures, obtained from β-thalassemia patients, confirming their activity as HbF inducers, and suggesting these molecules as lead compounds for further chemical and biological investigations.
Collapse
Affiliation(s)
- Giulia Breveglieri
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Salvatore Pacifico
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.P.); (D.P.); (C.T.)
| | - Cristina Zuccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Shaiq Sultan
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Elisabetta D’Aversa
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
| | - Delia Preti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.P.); (D.P.); (C.T.)
| | - Claudio Trapella
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.P.); (D.P.); (C.T.)
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.P.); (D.P.); (C.T.)
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy; (G.B.); (C.Z.); (L.C.C.); (S.S.); (E.D.); (R.G.)
- Center of Biotechnology, University of Ferrara, Via Fossato di Mortara 64b, 44121 Ferrara, Italy
| |
Collapse
|
7
|
Bosquesi PL, Melchior ACB, Pavan AR, Lanaro C, de Souza CM, Rusinova R, Chelucci RC, Barbieri KP, Fernandes GFDS, Carlos IZ, Andersen OS, Costa FF, Dos Santos JL. Synthesis and evaluation of resveratrol derivatives as fetal hemoglobin inducers. Bioorg Chem 2020; 100:103948. [PMID: 32450391 PMCID: PMC8052979 DOI: 10.1016/j.bioorg.2020.103948] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/29/2022]
Abstract
Resveratrol (RVT) derivatives (10a-i) were designed, synthesized, and evaluated for their potential as gamma-globin inducers in treating Sickle Cell Disease (SCD) symptoms. All compounds were able to release NO at different levels ranging from 0 to 26.3%, while RVT did not demonstrate this effect. In vivo, the antinociceptive effect was characterized using an acetic acid-induced abdominal contortion model. All compounds exhibited different levels of protection, ranging from 5.9 to 37.3%; the compound 10a was the most potent among the series. At concentrations between 3.13 and 12.5 µM, the derivative 10a resulted in a reduction of 41.1-64.3% in the TNF-α levels in the supernatants of macrophages that were previously LPS-stimulated. This inhibitory effect was higher than that of RVT used as the control. In addition, the compound 10a and RVT induced double the production of the gamma-globin chains (γG + γA), compared to the vehicle, using CD34+ cells. Compound 10a also did not induce membrane perturbation and it was not mutagenic in the in vivo assay. Thus, compound 10a emerged as a new prototype of the gamma-globin-inducer group with additional analgesic and anti-inflammatory activities and proving to be a useful alternative to treat SCD symptoms.
Collapse
Affiliation(s)
| | | | - Aline Renata Pavan
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Carolina Lanaro
- University of Campinas (UNICAMP), Hematology and Hemotherapy Center, Campinas 13083-878, Brazil
| | | | - Radda Rusinova
- Weill Cornell Medical College, Department of Physiology and Biophysics, New York, NY 10065-489, United States
| | - Rafael Consolin Chelucci
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Karina Pereira Barbieri
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | | | - Iracilda Zepone Carlos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Olaf Sparre Andersen
- Weill Cornell Medical College, Department of Physiology and Biophysics, New York, NY 10065-489, United States
| | - Fernando Ferreira Costa
- University of Campinas (UNICAMP), Hematology and Hemotherapy Center, Campinas 13083-878, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil.
| |
Collapse
|
8
|
Guo L, Chen J, Wang Q, Zhang J, Huang W. Oridonin enhances γ‑globin expression in erythroid precursors from patients with β‑thalassemia via activation of p38 MAPK signaling. Mol Med Rep 2019; 21:909-917. [PMID: 31789406 DOI: 10.3892/mmr.2019.10848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 09/20/2019] [Indexed: 11/05/2022] Open
Abstract
Upregulation of fetal hemoglobin expression can alleviate the severity of β‑thalassaemia. This study aimed to investigate the effects of Oridonin (ORI, a diterpenoid compound) on γ‑globin expression in human erythroid precursor cells and the potential underlying mechanisms. Erythroid precursor cells were enriched from 12 patients with β‑thalassaemia by two‑phase culture. The cells were then treated with different doses of ORI and the survival of erythroid precursor cells was determined. In addition, the expression levels of γ‑globin and potential mechanisms were analyzed by reverse transcription‑quantitative PCR, western blotting and chromatin immunoprecipitation. Treatment with 0.5 µM ORI preferably enhanced γ‑globin expression and exhibited little cytotoxicity. Similar to sodium butyrate (NaB, a histone deacetylase inhibitor), ORI significantly increased p38 mitogen‑activated protein kinase (MAPK) activation, γ‑globin expression, histone H3 and H4 acetylation at the Gγ‑ and Aγ‑globin promoters, and cAMP‑response element binding protein 1 (CREB1) phosphorylation. These effects were significantly mitigated by treatment with SB23580, a p38 MAPK inhibitor, in erythroid precursor cells. Therefore, ORI may effectively enhance γ‑globin expression by activating p38 MAPK and CREB1, leading to histone modification in γ‑globin gene promoters during the maturation of erythroid precursor cells. These findings suggested that ORI may be a novel and potential therapeutic agent for the treatment of β‑thalassaemia.
Collapse
Affiliation(s)
- Lishan Guo
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jia Chen
- Department of Neonatology, Pediatric Clinics of Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Qianying Wang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Junliang Zhang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Weimin Huang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
9
|
Liu L, Zhu X, Yu A, Ward CM, Pace BS. δ-Aminolevulinate induces fetal hemoglobin expression by enhancing cellular heme biosynthesis. Exp Biol Med (Maywood) 2019; 244:1220-1232. [PMID: 31475864 DOI: 10.1177/1535370219872995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Sickle cell disease (SCD) and β-thalassemia are inherited blood disorders caused by genetic defects in the β-globin gene on chromosome 11, producing severe disease in people worldwide. Induction of fetal hemoglobin consisting of two α-globin and two γ-globin chains ameliorates the clinical symptoms of both disorders. In the present study, we investigated the ability of δ-aminolevulinate (ALA), the heme precursor, to activate γ-globin gene expression as well as its effects on cellular functions in erythroid cell systems. We demonstrated that ALA induced γ-globin expression at both the transcriptional and protein levels in the KU812 erythroid cell line. Using inhibitors targeting two enzymes in the heme biosynthesis pathway, we showed that cellular heme biosynthesis was involved in ALA-mediated γ-globin activation. Moreover, the transcription factor NRF2 (nuclear factor [erythroid-derived 2]-like 2), a critical regulator of the cellular antioxidant response, was activated by ALA and contributed to mechanisms of γ-globin activation; ALA did not affect cell proliferation and was not toxic to cells. Subsequent studies demonstrated ALA-induced γ-globin activation in erythroid progenitors generated from normal human CD34+ stem cells. These data support future study to explore the potential of stimulating intracellular heme biosynthesis by ALA or similar compounds as a novel therapeutic strategy for treating SCD and β-thalassemia. Impact statement Inherited mutations in the β-globin-like genes result in the most common forms of genetic blood disease including sickle cell disease (SCD) and β-thalassemia worldwide. Therefore, effective inexpensive therapies that can be distributed widely are highly desirable. Currently, drug-mediated fetal hemoglobin (HbF) induction can ameliorate clinical symptoms of SCD and β-thalassemia and is the most effective strategy for developing new therapeutic options. In the current study, we confirmed that δ-Aminolevulinate (ALA), the precursor of heme, induces γ-globin expression at both the transcriptional and translational levels in primary human erythroid progenitors. Moreover, the results indicate activation of the transcription factor NRF2 (nuclear factor (erythroid-derived 2)-like 2) by ALA to enhance HbF expression. These data support future study to explore the potential of stimulating intracellular heme biosynthesis by ALA or similar compounds as a novel therapeutic strategy for treating SCD and β-thalassemia.
Collapse
Affiliation(s)
- Li Liu
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Xingguo Zhu
- Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Alexander Yu
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Christina M Ward
- Department of Biochemistry and Molecular Biology, Boston University, Boston, MA 02118, USA
| | - Betty S Pace
- Department of Pediatrics, Augusta University, Augusta, GA 30912, USA.,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
10
|
Das SS, Sinha R, Chakravorty N. Integrative microRNA and gene expression analysis identifies new drug repurposing candidates for fetal hemoglobin induction in β-hemoglobinopathies. Gene 2019; 706:77-83. [PMID: 31048070 DOI: 10.1016/j.gene.2019.04.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/18/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic induction of fetal hemoglobin (HbF) is one of the most promising approaches to ameliorate the severity of hemoglobinopathies like β-thalassemia and sickle cell anemia. Although several pharmacological agents have been investigated for HbF induction in adults, the majority of these are associated with significant side-effects. While drug repurposing is known to open new doors for the use of approved drugs in unexplored clinical conditions, the primary challenge lies in identifying such candidates. In this study, we aimed to identify repurposing candidates for HbF induction using a novel in silico approach utilizing microRNA-pathway-drug relationships. A computational drug repurposing strategy identified several unique candidates for HbF induction; among which Curcumin, Ginsenoside, Valproate, and Vorinostat were found to be most suitable for future trials. This study identified new drug repurposing candidates for HbF induction and demonstrates an easily adaptable methodology that can be used for other pathophysiological conditions.
Collapse
Affiliation(s)
- Sankha Subhra Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| | - Rashmi Sinha
- B. C. Roy Technology Hospital, Indian Institute of Technology Kharagpur, West Bengal 721302, India; Plant Hospital, Bharatiya Reserve Bank Note Mudran Private Limited (BRBNMPL), Salboni, West Bengal 721132, India
| | - Nishant Chakravorty
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
11
|
Robust erythroid differentiation system for rhesus hematopoietic progenitor cells allowing preclinical screening of genetic treatment strategies for the hemoglobinopathies. Cytotherapy 2018; 20:1278-1287. [PMID: 30249524 DOI: 10.1016/j.jcyt.2018.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS γ-globin expression can be induced by various gene modification strategies, which could be beneficial for hemoglobin (Hb) disorders. To translate promising ideas into clinics, large animal models have proven valuable to evaluate safety and efficacy of the approaches; however, in vitro erythroid differentiation methods have not been established to determine whether they can be modeled in nonhuman primates. METHODS We optimized erythroid differentiation culture to produce high-level adult Hb from rhesus hematopoietic progenitor cells by using low (LC) or high cytokine concentration (HC) protocols with or without feeder cells. In addition, we established rhesus globin protein analysis using reverse-phase high performance liquid chromatography and mass spectrometry. RESULTS Robust adult Hb production at protein levels was observed in the LC protocol when feeder cells were used, whereas the HC protocol resulted in higher baseline fetal Hb levels (P < 0.01). We then compared lentiviral transduction of rhesus cells between serum-containing LC media and serum-free StemSpan-based differentiation media, revealing 100-fold more efficient transduction in serum-free differentiation media (P < 0.01). Finally, rhesus CD34+ cells were transduced with lentiviral vectors encoding artificial zinc finger proteins (ZF-Ldb1), which can reactivate γ-globin expression via tethering the transcriptional co-regulator Ldb1 to γ-globin promoters, and were differentiated in the optimized erythroid differentiation method. This resulted in marked increases of γ-globin levels compared with control groups (P < 0.01). DISCUSSION In conclusion, we developed an efficient rhesus erythroid differentiation protocol from hematopoietic progenitor cells with low fetal and high adult Hb production. Further studies are warranted to optimize gene modification and transplantation of rhesus hematopoietic progenitor cells.
Collapse
|
12
|
Feriotto G, Marchetti N, Costa V, Torricelli P, Beninati S, Tagliati F, Mischiati C. Selected terpenes from leaves of Ocimum basilicum L. induce hemoglobin accumulation in human K562 cells. Fitoterapia 2018; 127:173-178. [DOI: 10.1016/j.fitote.2018.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/07/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022]
|
13
|
Melo TRFD, Kumkhaek C, Fernandes GFDS, Lopes Pires ME, Chelucci RC, Barbieri KP, Coelho F, Capote TSDO, Lanaro C, Carlos IZ, Marcondes S, Chegaev K, Guglielmo S, Fruttero R, Chung MC, Costa FF, Rodgers GP, Dos Santos JL. Discovery of phenylsulfonylfuroxan derivatives as gamma globin inducers by histone acetylation. Eur J Med Chem 2018; 154:341-353. [PMID: 29852459 DOI: 10.1016/j.ejmech.2018.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/05/2018] [Accepted: 05/07/2018] [Indexed: 12/17/2022]
Abstract
N-oxide derivatives 5(a-b), 8(a-b), and 11(a-c) were designed, synthesized and evaluated in vitro and in vivo as potential drugs that are able to ameliorate sickle cell disease (SCD) symptoms. All of the compounds demonstrated the capacity to releasing nitric oxide at different levels ranging from 0.8 to 30.1%, in vivo analgesic activity and ability to reduce TNF-α levels in the supernatants of monocyte cultures. The most active compound (8b) protected 50.1% against acetic acid-induced abdominal constrictions, while dipyrone, which was used as a control only protected 35%. Compounds 8a and 8b inhibited ADP-induced platelet aggregation by 84% and 76.1%, respectively. Both compounds increased γ-globin in K562 cells at 100 μM. The mechanisms involved in the γ-globin increase are related to the acetylation of histones H3 and H4 that is induced by these compounds. In vitro, the most promising compound (8b) was not cytotoxic, mutagenic and genotoxic.
Collapse
Affiliation(s)
| | - Chutima Kumkhaek
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD 20892, United States
| | | | - Maria Elisa Lopes Pires
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas, 13083-970, Brazil
| | - Rafael Consolin Chelucci
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Karina Pereira Barbieri
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Fernanda Coelho
- São Paulo State University (UNESP), School of Dentistry, Araraquara, 14801-903, Brazil
| | | | - Carolina Lanaro
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas, 13083-970, Brazil
| | - Iracilda Zeppone Carlos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Sisi Marcondes
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas, 13083-970, Brazil
| | - Konstantin Chegaev
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, 10124, Italy
| | - Stefano Guglielmo
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, 10124, Italy
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, 10124, Italy
| | - Man Chin Chung
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas, 13083-970, Brazil
| | - Griffin P Rodgers
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD 20892, United States
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil.
| |
Collapse
|
14
|
Migliaccio AR, Varricchio L. Concise Review: Advanced Cell Culture Models for Diamond Blackfan Anemia and Other Erythroid Disorders. Stem Cells 2018; 36:172-179. [PMID: 29124822 PMCID: PMC5785423 DOI: 10.1002/stem.2735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/14/2017] [Accepted: 11/01/2017] [Indexed: 01/26/2023]
Abstract
In vitro surrogate models of human erythropoiesis made many contributions to our understanding of the extrinsic and intrinsic regulation of this process in vivo and how they are altered in erythroid disorders. In the past, variability among the levels of hemoglobin F produced by adult erythroblasts generated in vitro by different laboratories identified stage of maturation, fetal bovine serum, and accessory cells as "confounding factors," that is, parameters intrinsically wired in the experimental approach that bias the results observed. The discovery of these factors facilitated the identification of drugs that accelerate terminal maturation or activate specific signaling pathways for the treatment of hemoglobinopathies. It also inspired studies to understand how erythropoiesis is regulated by macrophages present in the erythroid islands. Recent cell culture advances have greatly increased the number of human erythroid cells that can be generated in vitro and are used as experimental models to study diseases, such as Diamond Blackfan Anemia, which were previously poorly amenable to investigation. However, in addition to the confounding factors already identified, improvement in the culture models has introduced novel confounding factors, such as possible interactions between signaling from cKIT, the receptor for stem cell factor, and from the glucocorticoid receptor, the cell proliferation potential and the clinical state of the patients. This review will illustrate these new confounding factors and discuss their clinical translation potential to improve our understanding of Diamond Blackfan Anemia and other erythroid disorders. Stem Cells 2018;36:172-179.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
| |
Collapse
|
15
|
Dai Y, Chen T, Ijaz H, Cho EH, Steinberg MH. SIRT1 activates the expression of fetal hemoglobin genes. Am J Hematol 2017; 92:1177-1186. [PMID: 28776729 DOI: 10.1002/ajh.24879] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023]
Abstract
High fetal hemoglobin (HbF, α2 γ2 ) levels ameliorate the clinical manifestations of sickle cell disease and β thalassemia. The mechanisms that repress HbF expression and silence γ-globin genes in adults are incompletely characterized and only a single HbF inducer, hydroxyurea, is approved for treatment, and only in patients with sickle cell disease. We identified SIRT1, a protein deacetylase, as a new inducer of γ-globin. SIRT1 knockdown decreased, while SIRT1 ectopic expression upregulated γ-globin gene (HBG) expression in primary human erythroid cells and in K562 cells. The small molecule SIRT1 activators SRT2104 and SRT1720 enhanced HBG expression in cord blood human erythroblasts and reactivated silenced HBG in adult human erythroblasts. Furthermore, SIRT1 binds in the β-globin gene cluster locus control region (LCR) and HBG promoters, promotes the looping of the LCR to HBG promoter, and increases the binding of RNA polymerase II and H4K16Ac in the HBG promoter. SIRT1 suppressed the expression of the HBG suppressors BCL11A, KLF1, HDAC1 and HDAC2. Lastly, SIRT1 did not change the proliferation of human erythroid progenitor cells or the expression of differentiation marker CD235a. These data suggest that SIRT1 activates HBG expression through facilitating LCR looping to the HBG promoter, inhibiting the expression of transcriptional suppressors of HBG, and indirectly increasing histone acetylation in the HBG promoter. SIRT1 is a potential therapeutic target for γ-globin gene induction, and small molecule SIRT1 activators might serve as a lead compound for the development of new HbF inducers.
Collapse
Affiliation(s)
- Yan Dai
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Tyngwei Chen
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Heba Ijaz
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Elizabeth H. Cho
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Martin H. Steinberg
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| |
Collapse
|
16
|
Shang X, Xu X. Update in the genetics of thalassemia: What clinicians need to know. Best Pract Res Clin Obstet Gynaecol 2017; 39:3-15. [DOI: 10.1016/j.bpobgyn.2016.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 08/18/2016] [Accepted: 10/14/2016] [Indexed: 11/17/2022]
|
17
|
Khan SA, Damanhouri G, Ali A, Khan SA, Khan A, Bakillah A, Marouf S, Al Harbi G, Halawani SH, Makki A. Precipitating factors and targeted therapies in combating the perils of sickle cell disease--- A special nutritional consideration. Nutr Metab (Lond) 2016; 13:50. [PMID: 27508000 PMCID: PMC4977632 DOI: 10.1186/s12986-016-0109-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 07/22/2016] [Indexed: 01/19/2023] Open
Abstract
Nutritional research in sickle cell disease has been the focus in recent times owing to not only specific nutritional deficiencies, but also the improvements associated with less painful episodes. Though hydroxyurea remains the drug of choice, certain adverse health effects on long term supplementation makes room for researches of different compounds. Macro and micro nutrient deficiencies, along with vitamins, play an important role in not only meeting the calorific needs, but also reducing clinical complications and growth abnormalities. Symptoms of hyper protein metabolism, increased cell turnover, increased cardiac output, and appetite suppression due to enhanced cytokine production, might give us leads for better understanding of the mechanisms involved. Different nutritional approaches comprising of traditional herbal therapies, antioxidants, flavonoids, vitamins, minerals etc., reducing oxidative stress and blood aggregation, have been tried out to increase the health potential. Nutritional therapies may also serve complementary to the newer therapies using ozone, hematopoietic stem cell transplantation, antifungal medications, erythropoietin etc. Herein we try to present a holistic picture of the different patho-physiological mechanisms, and nutritional strategies adopted.
Collapse
Affiliation(s)
- Shahida A Khan
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ghazi Damanhouri
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ashraf Ali
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Sarah A Khan
- National Brain Research Center, Manesar, Gurgaon, 122051 India
| | - Aziz Khan
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ahmed Bakillah
- Department of Medicine, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, New York 11203 United State of America (USA)
| | - Samy Marouf
- Department of Hematology, King Fahd Hospital of the Armed forces, Jeddah, Kingdom of Saudi Arabia ; Department of Medical Laboratory, King Fahd Hospital of the Armed forces, Jeddah, Kingdom of Saudi Arabia
| | - Ghazi Al Harbi
- Department of Hematology, Soliman Fakeeh Hospital Jeddah, Jeddah, Kingdom of Saudi Arabia
| | - Saeed H Halawani
- Department of Hematology, Umm Al Qura University, Faculty of Medicine, Makkah, Kingdom of Saudi Arabia
| | - Ahmad Makki
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| |
Collapse
|
18
|
Tayebi B, Abrishami F, Alizadeh S, Minayi N, Mohammadian M, Soleimani M, Dehghanifard A, Atwan H, Ajami M, Ajami M. Modulation of microRNAs expression in hematopoietic stem cells treated with sodium butyrate in inducing fetal hemoglobin expression. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:146-156. [PMID: 26837891 DOI: 10.3109/21691401.2016.1138487] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Context Inherited hemoglobin diseases are the most common single-gene disorders. Induction of fetal hemoglobin in beta hemoglobin disorders compensate for abnormal chain and ameliorate the clinical complications. Sodium butyrate is used conventionally for fetal hemoglobin induction; it can be replaced by safer therapeutic tools like microRNAs, small non-coding RNAs that control number of epigenetic mechanisms. Objective In this study, we compared the changes in the microRNAs of differentiated erythroid cells between control and sodium butyrate treated groups. The objective is to find significant association between these changes and gamma chain up regulation. Materials and methods First, CD133+ hematopoietic stem cells were isolated from cord blood by magnetic cell sorting (MACS) technique. After proliferation, the cells were differentiated to erythroid lineage in culture medium by EPO, SCF, and IL3. Meanwhile, the test group was treated with sodium butyrate. Then, gamma chain upregulation was verified by qPCR technique. Finally, microRNA profiling was performed through microarray assay and some of them confirmed by qPCR. Result Results demonstrated that gamma chain was 5.9-fold upregulated in the treated group. Significant changes were observed at 76 microRNAs, in which 20 were up-regulated and 56 were down-regulated. Discussion Five of these microRNAs including U101, hsa-miR-4726-5p, hsa-miR7109 5p, hsa-miR3663, and hsa-miR940 had significant changes in expression and volume. Conclusion In conclusion, it can be assumed that sodium butyrate can up-regulate gamma chain gene, and change miRNAs expression. These results can be profitable in future studies to find therapeutic goal suitable for such disorders.
Collapse
Affiliation(s)
- Behnoosh Tayebi
- a Faculty of Medicine , Qaem Hospital, Mashhad University of Medical Sciences and Health Care Services , Mashhad , Iran
| | - Fatemeh Abrishami
- a Faculty of Medicine , Qaem Hospital, Mashhad University of Medical Sciences and Health Care Services , Mashhad , Iran
| | - Shaban Alizadeh
- b Hematology Department, School of Allied Medical , Tehran University of Medical Sciences , Tehran , Iran
| | - Neda Minayi
- b Hematology Department, School of Allied Medical , Tehran University of Medical Sciences , Tehran , Iran
| | - Mozhdeh Mohammadian
- c Amol Faculty of Paramedical Sciences , Mazandaran University of Medical Sciences , Sari , Iran.,d Hematology and Oncology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Masoud Soleimani
- e Department of Hematology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | - Ali Dehghanifard
- f Taleghani Bone Marrow Transplantation Center, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Hossein Atwan
- g Immunology Department , Iran University of Medical Science , Tehran , Iran
| | - Monireh Ajami
- a Faculty of Medicine , Qaem Hospital, Mashhad University of Medical Sciences and Health Care Services , Mashhad , Iran
| | - Mansoureh Ajami
- e Department of Hematology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
19
|
Dai Y, Sangerman J, Luo HY, Fucharoen S, Chui DHK, Faller DV, Perrine SP. Therapeutic fetal-globin inducers reduce transcriptional repression in hemoglobinopathy erythroid progenitors through distinct mechanisms. Blood Cells Mol Dis 2015; 56:62-9. [PMID: 26603726 DOI: 10.1016/j.bcmd.2015.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/26/2015] [Indexed: 12/15/2022]
Abstract
Pharmacologic augmentation of γ-globin expression sufficient to reduce anemia and clinical severity in patients with diverse hemoglobinopathies has been challenging. In studies here, representative molecules from four chemical classes, representing several distinct primary mechanisms of action, were investigated for effects on γ-globin transcriptional repressors, including components of the NuRD complex (LSD1 and HDACs 2-3), and the downstream repressor BCL11A, in erythroid progenitors from hemoglobinopathy patients. Two HDAC inhibitors (MS-275 and SB939), a short-chain fatty acid derivative (sodium dimethylbutyrate [SDMB]), and an agent identified in high-throughput screening, Benserazide, were studied. These therapeutics induced γ-globin mRNA in progenitors above same subject controls up to 20-fold, and increased F-reticulocytes up to 20%. Cellular protein levels of BCL11A, LSD-1, and KLF1 were suppressed by the compounds. Chromatin immunoprecipitation assays demonstrated a 3.6-fold reduction in LSD1 and HDAC3 occupancy in the γ-globin gene promoter with Benserazide exposure, 3-fold reduction in LSD-1 and HDAC2 occupancy in the γ-globin gene promoter with SDMB exposure, while markers of gene activation (histone H3K9 acetylation and H3K4 demethylation), were enriched 5.7-fold. These findings identify clinical-stage oral therapeutics which inhibit or displace major co-repressors of γ-globin gene transcription and may suggest a rationale for combination therapy to produce enhanced efficacy.
Collapse
Affiliation(s)
- Yan Dai
- Hemoglobinopathy Thalassemia Research Unit and Cancer Center, Boston University School of Medicine, Boston, MA, United States
| | - Jose Sangerman
- Hemoglobinopathy Thalassemia Research Unit and Cancer Center, Boston University School of Medicine, Boston, MA, United States
| | - Hong Yuan Luo
- Department of Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Suthat Fucharoen
- Thalassemia Research Center, Mahidol University, Phuttamonthon, Thailand
| | - David H K Chui
- Department of Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Douglas V Faller
- Hemoglobinopathy Thalassemia Research Unit and Cancer Center, Boston University School of Medicine, Boston, MA, United States; Phoenicia BioSciences, Inc., Newton, MA, United States
| | - Susan P Perrine
- Hemoglobinopathy Thalassemia Research Unit and Cancer Center, Boston University School of Medicine, Boston, MA, United States; Phoenicia BioSciences, Inc., Newton, MA, United States; Center for Hemoglobin Research in Minorities, Departments of Pediatrics and Medicine, Howard University College of Medicine, Washington DC, United States.
| |
Collapse
|