1
|
Yasara N, Premawardhena A, Mettananda S. A comprehensive review of hydroxyurea for β-haemoglobinopathies: the role revisited during COVID-19 pandemic. Orphanet J Rare Dis 2021; 16:114. [PMID: 33648529 PMCID: PMC7919989 DOI: 10.1186/s13023-021-01757-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background Hydroxyurea is one of the earliest drugs that showed promise in the management of haemoglobinopathies that include β-thalassaemia and sickle cell disease. Despite this, many aspects of hydroxyurea are either unknown or understudied; specifically, its usefulness in β-thalassaemia major and haemoglobin E β-thalassaemia is unclear. However, during COVID-19 pandemic, it has become a valuable adjunct to transfusion therapy in patients with β-haemoglobinopathies. In this review, we aim to explore the available in vitro and in vivo mechanistic data and the clinical utility of hydroxyurea in β-haemoglobinopathies with a special emphasis on its usefulness during the COVID-19 pandemic. Main body Hydroxyurea is an S-phase-specific drug that reversibly inhibits ribonucleoside diphosphate reductase enzyme which catalyses an essential step in the DNA biosynthesis. In human erythroid cells, it induces the expression of γ-globin, a fetal globin gene that is suppressed after birth. Through several molecular pathways described in this review, hydroxyurea exerts many favourable effects on the haemoglobin content, red blood cell indices, ineffective erythropoiesis, and blood rheology in patients with β-haemoglobinopathies. Currently, it is recommended for sickle cell disease and non-transfusion dependent β-thalassaemia. A number of clinical trials are ongoing to evaluate its usefulness in transfusion dependent β-thalassaemia. During the COVID-19 pandemic, it was widely used as an adjunct to transfusion therapy due to limitations in the availability of blood and logistical disturbances. Thus, it has become clear that hydroxyurea could play a remarkable role in reducing transfusion requirements of patients with haemoglobinopathies, especially when donor blood is a limited resource. Conclusion Hydroxyurea is a well-tolerated oral drug which has been in use for many decades. Through its actions of reversible inhibition of ribonucleoside diphosphate reductase enzyme and fetal haemoglobin induction, it exerts many favourable effects on patients with β-haemoglobinopathies. It is currently approved for the treatment of sickle cell disease and non-transfusion dependent β-thalassaemia. Also, there are various observations to suggest that hydroxyurea is an important adjunct in the treatment of transfusion dependent β-thalassaemia which should be confirmed by randomised clinical trials.
Collapse
Affiliation(s)
- Nirmani Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
| | - Anuja Premawardhena
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka.,Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - Sachith Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka. .,Colombo North Teaching Hospital, Ragama, Sri Lanka.
| |
Collapse
|
2
|
Pace BS, Starlard-Davenport A, Kutlar A. Sickle cell disease: progress towards combination drug therapy. Br J Haematol 2021; 194:240-251. [PMID: 33471938 DOI: 10.1111/bjh.17312] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022]
Abstract
Dr. John Herrick described the first clinical case of sickle cell anaemia (SCA) in the United States in 1910. Subsequently, four decades later, Ingram and colleagues characterized the A to T substitution in DNA producing the GAG to GTG codon and replacement of glutamic acid with valine in the sixth position of the βS -globin chain. The establishment of Comprehensive Sickle Cell Centers in the United States in the 1970s was an important milestone in the development of treatment strategies and describing the natural history of sickle cell disease (SCD) comprised of genotypes including homozygous haemoglobin SS (HbSS), HbSβ0 thalassaemia, HbSC and HbSβ+ thalassaemia, among others. Early drug studies demonstrating effective treatments of HbSS and HbSβ0 thalassaemia, stimulated clinical trials to develop disease-specific therapies to induce fetal haemoglobin due to its ability to block HbS polymerization. Subsequently, hydroxycarbamide proved efficacious in adults with SCA and was Food and Drug Administration (FDA)-approved in 1998. After two decades of hydroxycarbamide use for SCD, there continues to be limited clinical acceptance of this chemotherapy drug, providing the impetus for investigators and pharmaceutical companies to develop non-chemotherapy agents. Investigative efforts to determine the role of events downstream of deoxy-HbS polymerization, such as endothelial cell activation, cellular adhesion, chronic inflammation, intravascular haemolysis and nitric oxide scavenging, have expanded drug targets which reverse the pathophysiology of SCD. After two decades of slow progress in the field, since 2018 three new drugs were FDA-approved for SCA, but research efforts to develop treatments continue. Currently over 30 treatment intervention trials are in progress to investigate a wide range of agents acting by complementary mechanisms, providing the rationale for ushering in the age of effective and safe combination drug therapy for SCD. Parallel efforts to develop curative therapies using haematopoietic stem cell transplant and gene therapy provide individuals with SCD multiple treatment options. We will discuss progress made towards drug development and potential combination drug therapy for SCD with the standard of care hydroxycarbamide.
Collapse
Affiliation(s)
- Betty S Pace
- Department of Pediatrics, Augusta University, Augusta, GA, USA.,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Athena Starlard-Davenport
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Abdullah Kutlar
- Department of Medicine, Center for Blood Disorders, Augusta University, Augusta, GA, USA
| |
Collapse
|
3
|
Ferreira WA, Chweih H, Lanaro C, Almeida CB, Brito PL, Gotardo EMF, Torres L, Miguel LI, Franco-Penteado CF, Leonardo FC, Garcia F, Saad STO, Frenette PS, Brockschnieder D, Costa FF, Stasch JP, Sandner P, Conran N. Beneficial Effects of Soluble Guanylyl Cyclase Stimulation and Activation in Sickle Cell Disease Are Amplified by Hydroxyurea: In Vitro and In Vivo Studies. J Pharmacol Exp Ther 2020; 374:469-478. [PMID: 32631869 PMCID: PMC7445859 DOI: 10.1124/jpet.119.264606] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
The complex pathophysiology of sickle cell anemia (SCA) involves intravascular hemolytic processes and recurrent vaso-occlusion, driven by chronic vascular inflammation, which result in the disease's severe clinical complications, including recurrent painful vaso-occlusive episodes. Hydroxyurea, the only drug frequently used for SCA therapy, is a cytostatic agent, although it appears to exert nitric oxide/soluble guanylyl cyclase (sGC) modulating activity. As new drugs that can complement or replace the use of hydroxyurea are sought to further reduce vaso-occlusive episode frequency in SCA, we investigated the effects of the sGC agonists BAY 60-2770 (sGC activator) and BAY 41-2272 (sGC stimulator) in the presence or absence of hydroxyurea on SCA vaso-occlusive mechanisms and cell recruitment both ex vivo and in vivo. These agents significantly reduced stimulated human SCA neutrophil adhesive properties ex vivo in association with the inhibition of surface β2-integrin activation. A single administration of BAY 60-2770 or BAY 41-2272 decreased tumor necrosis factor cytokine-induced leukocyte recruitment in a mouse model of SCA vaso-occlusion. Importantly, the in vivo actions of both agonists were significantly potentiated by the coadministration of hydroxyurea. Erythroid cell fetal hemoglobin (HbF) elevation is also a major goal for SCA therapy. BAY 41-2272 but not BAY 60-2770 at the concentrations employed significantly induced γ-globin gene transcription in association with HbF production in cultured erythroleukemic cells. In conclusion, sGC agonist drugs could represent a promising approach as therapy for SCA, for use either as stand-alone treatments or in combination with hydroxyurea. SIGNIFICANCE STATEMENT: This preclinical study demonstrates that stimulators and activators of sGC are potent inhibitors of the adhesion and recruitment of leukocytes from humans and in mice with sickle cell anemia (SCA) and may represent a promising approach for diminishing vaso-occlusive episode frequency in SCA. Hydroxyurea, a drug already frequently used for treating SCA, was found to potentiate the beneficial effects of sGC agonists in in vivo studies, implying that these classes of compounds could be used alone or in combination therapy.
Collapse
Affiliation(s)
- W A Ferreira
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - H Chweih
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - C Lanaro
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - C B Almeida
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - P L Brito
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - E M F Gotardo
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - L Torres
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - L I Miguel
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - C F Franco-Penteado
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - F C Leonardo
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - F Garcia
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - S T O Saad
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - P S Frenette
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - D Brockschnieder
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - F F Costa
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - J P Stasch
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - P Sandner
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| | - N Conran
- Hematology Center, School of Medical Sciences, University of Campinas (UNICAMP), Brazil (W.A.F., H.C., C.L., C.B.A., P.L.B., E.M.F.G., L.T., L.I.M., C.F.F.-P., F.C.L., F.G., S.S.T.O., F.F.C., N.C.); Bayer AG, Pharmaceuticals - Drug Discovery, Wuppertal, Germany (D.B., J.P.S., P.S.); Ruth L. and David S Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York (P.S.F.); and Hannover Medical School, Institute of Pharmacology, Hannover, Germany (P.S.)
| |
Collapse
|
4
|
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
|
5
|
Stephanou C, Tamana S, Minaidou A, Papasavva P, Kleanthous M, Kountouris P. Genetic Modifiers at the Crossroads of Personalised Medicine for Haemoglobinopathies. J Clin Med 2019; 8:E1927. [PMID: 31717530 PMCID: PMC6912721 DOI: 10.3390/jcm8111927] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/25/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
Haemoglobinopathies are common monogenic disorders with diverse clinical manifestations, partly attributed to the influence of modifier genes. Recent years have seen enormous growth in the amount of genetic data, instigating the need for ranking methods to identify candidate genes with strong modifying effects. Here, we present the first evidence-based gene ranking metric (IthaScore) for haemoglobinopathy-specific phenotypes by utilising curated data in the IthaGenes database. IthaScore successfully reflects current knowledge for well-established disease modifiers, while it can be dynamically updated with emerging evidence. Protein-protein interaction (PPI) network analysis and functional enrichment analysis were employed to identify new potential disease modifiers and to evaluate the biological profiles of selected phenotypes. The most relevant gene ontology (GO) and pathway gene annotations for (a) haemoglobin (Hb) F levels/Hb F response to hydroxyurea included urea cycle, arginine metabolism and vascular endothelial growth factor receptor (VEGFR) signalling, (b) response to iron chelators included xenobiotic metabolism and glucuronidation, and (c) stroke included cytokine signalling and inflammatory reactions. Our findings demonstrate the capacity of IthaGenes, together with dynamic gene ranking, to expand knowledge on the genetic and molecular basis of phenotypic variation in haemoglobinopathies and to identify additional candidate genes to potentially inform and improve diagnosis, prognosis and therapeutic management.
Collapse
Affiliation(s)
| | | | | | | | - Marina Kleanthous
- Correspondence: (M.K.); (P.K.); Tel.:+357-2239-2652 (M.K.); +357-2239-2623 (P.K.)
| | - Petros Kountouris
- Correspondence: (M.K.); (P.K.); Tel.:+357-2239-2652 (M.K.); +357-2239-2623 (P.K.)
| |
Collapse
|
6
|
Abstract
IMPACT STATEMENT Sickle cell disease (SCD) is one of the most common inherited diseases and is associated with a reduced life expectancy and acute and chronic complications, including frequent painful vaso-occlusive episodes that often require hospitalization. At present, treatment of SCD is limited to hematopoietic stem cell transplant, transfusion, and limited options for pharmacotherapy, based principally on hydroxyurea therapy. This review highlights the importance of intracellular cGMP-dependent signaling pathways in SCD pathophysiology; modulation of these pathways with soluble guanylate cyclase (sGC) stimulators or phosphodiesterase (PDE) inhibitors could potentially provide vasorelaxation and anti-inflammatory effects, as well as elevate levels of anti-sickling fetal hemoglobin.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas – UNICAMP,
Cidade Universitária, Campinas-SP 13083-878-SP, Brazil
| | - Lidiane Torres
- Hematology Center, University of Campinas – UNICAMP,
Cidade Universitária, Campinas-SP 13083-878-SP, Brazil
| |
Collapse
|
7
|
Ikuta T, Sellak H, Liu SY, Odo N. Serum of sickle cell disease patients contains fetal hemoglobin silencing factors secreted from leukocytes. J Blood Med 2018; 9:95-104. [PMID: 29950916 PMCID: PMC6018840 DOI: 10.2147/jbm.s156999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The mechanisms that regulate fetal hemoglobin (HbF) expression in sickle cell disease (SCD) remain elusive. We previously showed that steady-state SCD patients with high HbF levels due to a γ-globin gene mutation demonstrate strong inverse correlations between HbF levels and leukocyte counts, suggesting that leukocytes play a role in regulating HbF in SCD. MATERIALS AND METHODS To further investigate the role of leukocytes in HbF expression in SCD, we examined the presence of HbF silencing factors in the serum of 82 SCD patients who received hydroxyurea (HU) therapy. RESULTS HU-mediated HbF induction was associated with elevated total hemoglobin levels and improved red blood cell parameters, but there was no correlation with reticulocyte or platelet counts. Importantly, we again found that HU-induced HbF levels correlated with reductions in both neutrophils and lymphocytes/monocytes, indicating that these cell lineages may have a role in regulating HU-mediated HbF expression. Our in vitro studies using CD34+-derived primary erythroblasts found that patient serum preparations include HbF silencing factors that are distinct from granulocyte-macrophage colony-stimulating factor, and the activity of such factors decreases upon HU therapy. CONCLUSION Together, these results demonstrate the importance of leukocyte numbers in the regulation of HbF levels for SCD patients both in steady state and under HU therapy, and that leukocytes secrete HbF silencing factors that negatively affect HbF expression in erythroid-lineage cells in SCD.
Collapse
Affiliation(s)
- Tohru Ikuta
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hassan Sellak
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Si-Yang Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nadine Odo
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
8
|
Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
9
|
Ghanta M, Panchanathan E, Lakkakula BVKS. Cyclic Guanosine Monophosphate-Dependent Protein Kinase I Stimulators and Activators Are Therapeutic Alternatives for Sickle Cell Disease. Turk J Haematol 2018; 35:77-78. [PMID: 29192603 PMCID: PMC5843781 DOI: 10.4274/tjh.2017.0407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 12/01/2017] [Indexed: 12/19/2022] Open
Affiliation(s)
- Mohankrishna Ghanta
- Sri Ramachandra Medical College and Research Institute-DU, Faculty of Medicine, Department of Pharmacology, Chennai, Tamil Nadu, India
| | - Elango Panchanathan
- Sri Ramachandra Medical College and Research Institute-DU, Faculty of Medicine, Department of Pharmacology, Chennai, Tamil Nadu, India
| | - Bhaskar VKS Lakkakula
- Sickle Cell Institute Chhattisgarh, Department of Molecular Genetics, Division of Research, Raipur, Chhattisgarh, India
| |
Collapse
|
10
|
Zhu X, Hu T, Ho MH, Wang Y, Yu M, Patel N, Pi W, Choi JH, Xu H, Ganapathy V, Kutlar F, Kutlar A, Tuan D. Hydroxyurea differentially modulates activator and repressors of γ-globin gene in erythroblasts of responsive and non-responsive patients with sickle cell disease in correlation with Index of Hydroxyurea Responsiveness. Haematologica 2017; 102:1995-2004. [PMID: 28971909 PMCID: PMC5709098 DOI: 10.3324/haematol.2017.175646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/29/2017] [Indexed: 01/24/2023] Open
Abstract
Hydroxyurea (HU), the first of two drugs approved by the US Food and Drug Administration for treating patients with sickle cell disease (SCD), produces anti-sickling effect by re-activating fetal γ-globin gene to enhance production of fetal hemoglobin. However, approximately 30% of the patients do not respond to HU therapy. The molecular basis of non-responsiveness to HU is not clearly understood. To address this question, we examined HU-induced changes in the RNA and protein levels of transcription factors NF-Y, GATA-1, -2, BCL11A, TR4, MYB and NF-E4 that assemble the γ-globin promoter complex and regulate transcription of γ-globin gene. In erythroblasts cultured from peripheral blood CD34+ cells of patients with SCD, we found that HU-induced changes in the protein but not the RNA levels of activator GATA-2 and repressors GATA-1, BCL11A and TR4 correlated with HU-induced changes in fetal hemoglobin (HbF) levels in the peripheral blood of HU high and low responders. However, HU did not significantly induce changes in the protein or RNA levels of activators NF-Y and NF-E4. Based on HU-induced changes in the protein levels of GATA-2, -1 and BCL11A, we calculated an Index of Hydroxyurea Responsiveness (IndexHU-3). Compared to the HU-induced fold changes in the individual transcription factor protein levels, the numerical values of IndexHU-3 statistically correlated best with the HU-induced peripheral blood HbF levels of the patients. Thus, IndexHU-3 can serve as an appropriate indicator for inherent HU responsiveness of patients with SCD.
Collapse
Affiliation(s)
- Xingguo Zhu
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| | - Tianxiang Hu
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| | - Meng Hsuan Ho
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,School of Dentistry, Meharry Medical College, Nashville, TN, USA
| | - Yongchao Wang
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Miao Yu
- Georgia Cancer Research Center, Augusta University, GA, USA
| | - Niren Patel
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Wenhu Pi
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeong-Hyeon Choi
- Georgia Cancer Research Center, Augusta University, GA, USA.,Department of Biostatistics, Augusta University, GA, USA
| | - Hongyan Xu
- Department of Biostatistics, Augusta University, GA, USA
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ferdane Kutlar
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Dorothy Tuan
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| |
Collapse
|
11
|
Cortese-Krott MM, Mergia E, Kramer CM, Lückstädt W, Yang J, Wolff G, Panknin C, Bracht T, Sitek B, Pernow J, Stasch JP, Feelisch M, Koesling D, Kelm M. Identification of a soluble guanylate cyclase in RBCs: preserved activity in patients with coronary artery disease. Redox Biol 2017; 14:328-337. [PMID: 29024896 PMCID: PMC5975213 DOI: 10.1016/j.redox.2017.08.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/21/2022] Open
Abstract
Endothelial dysfunction is associated with decreased NO bioavailability and impaired activation of the NO receptor soluble guanylate cyclase (sGC) in the vasculature and in platelets. Red blood cells (RBCs) are known to produce NO under hypoxic and normoxic conditions; however evidence of expression and/or activity of sGC and downstream signaling pathway including phopshodiesterase (PDE)-5 and protein kinase G (PKG) in RBCs is still controversial. In the present study, we aimed to investigate whether RBCs carry a functional sGC signaling pathway and to address whether this pathway is compromised in coronary artery disease (CAD). Using two independent chromatographic procedures, we here demonstrate that human and murine RBCs carry a catalytically active α1β1-sGC (isoform 1), which converts 32P-GTP into 32P-cGMP, as well as PDE5 and PKG. Specific sGC stimulation by NO+BAY 41-2272 increases intracellular cGMP-levels up to 1000-fold with concomitant activation of the canonical PKG/VASP-signaling pathway. This response to NO is blunted in α1-sGC knockout (KO) RBCs, but fully preserved in α2-sGC KO. In patients with stable CAD and endothelial dysfunction red cell eNOS expression is decreased as compared to aged-matched controls; by contrast, red cell sGC expression/activity and responsiveness to NO are fully preserved, although sGC oxidation is increased in both groups. Collectively, our data demonstrate that an intact sGC/PDE5/PKG-dependent signaling pathway exists in RBCs, which remains fully responsive to NO and sGC stimulators/activators in patients with endothelial dysfunction. Targeting this pathway may be helpful in diseases with NO deficiency in the microcirculation like sickle cell anemia, pulmonary hypertension, and heart failure.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany.
| | - Evanthia Mergia
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Jiangning Yang
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Karolinska Universitetssjukhuset, Solna, 171 76 Stockholm, Sweden
| | - Georg Wolff
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Christina Panknin
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Thilo Bracht
- Medizinisches Proteom-Center, Ruhr- University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr- University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - John Pernow
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Karolinska Universitetssjukhuset, Solna, 171 76 Stockholm, Sweden
| | - Johannes-Peter Stasch
- Bayer Pharma AG, Aprather Weg 18a, 42096 Wuppertal, Germany; Institute of Pharmacy, University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle (Saale), Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Tremona Road, SO166YD Southampton, United Kingdom
| | - Doris Koesling
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| |
Collapse
|
12
|
Wang WC. Minireview: Prognostic factors and the response to hydroxurea treatment in sickle cell disease. Exp Biol Med (Maywood) 2016; 241:730-6. [PMID: 27026724 DOI: 10.1177/1535370216642048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This review describes current considerations in the use of hydroxyurea for the management of sickle cell disease in the context of clinical severity. Randomized trials of hydroxyurea have generally enrolled subjects with increased severity based on frequent vaso-occlusive events. An exception was the BABY HUG study in infants which documented substantial benefit even for asymptomatic subjects. Increasing data indicate that hydroxyurea has a substantial effect on reducing mortality in both adults and children-perhaps the most compelling reason for advocating the drug's widespread use. Although the efficacy of hydroxyurea is mediated primarily through increased erythrocyte fetal hemoglobin and much has been learned about the genomic influences on fetal hemoglobin levels in sickle cell disease, our ability to predict the fetal hemoglobin response to hydroxyurea remains limited; much more work in this area is indicated. The review is concluded with the recommendations of the 2014 NIH Evidence-Based Management of Sickle Cell Disease Expert Panel Report.
Collapse
Affiliation(s)
- Winfred C Wang
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
13
|
Nitric Oxide-cGMP Signaling Stimulates Erythropoiesis through Multiple Lineage-Specific Transcription Factors: Clinical Implications and a Novel Target for Erythropoiesis. PLoS One 2016; 11:e0144561. [PMID: 26727002 PMCID: PMC4699757 DOI: 10.1371/journal.pone.0144561] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/19/2015] [Indexed: 11/19/2022] Open
Abstract
Much attention has been directed to the physiological effects of nitric oxide (NO)-cGMP signaling, but virtually nothing is known about its hematologic effects. We reported for the first time that cGMP signaling induces human γ-globin gene expression. Aiming at developing novel therapeutics for anemia, we examined here the hematologic effects of NO-cGMP signaling in vivo and in vitro. We treated wild-type mice with NO to activate soluble guanylate cyclase (sGC), a key enzyme of cGMP signaling. Compared to untreated mice, NO-treated mice had higher red blood cell counts and total hemoglobin but reduced leukocyte counts, demonstrating that when activated, NO-cGMP signaling exerts hematopoietic effects on multiple types of blood cells in vivo. We next generated mice which overexpressed rat sGC in erythroid and myeloid cells. The forced expression of sGCs activated cGMP signaling in both lineage cells. Compared with non-transgenic littermates, sGC mice exhibited hematologic changes similar to those of NO-treated mice. Consistently, a membrane-permeable cGMP enhanced the differentiation of hematopoietic progenitors toward erythroid-lineage cells but inhibited them toward myeloid-lineage cells by controlling multiple lineage-specific transcription factors. Human γ-globin gene expression was induced at low but appreciable levels in sGC mice carrying the human β-globin locus. Together, these results demonstrate that NO-cGMP signaling is capable of stimulating erythropoiesis in both in vitro and vivo settings by controlling the expression of multiple lineage-specific transcription factors, suggesting that cGMP signaling upregulates erythropoiesis at the level of gene transcription. The NO-cGMP signaling axis may constitute a novel target to stimulate erythropoiesis in vivo.
Collapse
|
14
|
Pace BS, Liu L, Li B, Makala LH. Cell signaling pathways involved in drug-mediated fetal hemoglobin induction: Strategies to treat sickle cell disease. Exp Biol Med (Maywood) 2015; 240:1050-64. [PMID: 26283707 DOI: 10.1177/1535370215596859] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The developmental regulation of globin gene expression has shaped research efforts to establish therapeutic modalities for individuals affected with sickle cell disease and β-thalassemia. Fetal hemoglobin has been shown to block sickle hemoglobin S polymerization to improve symptoms of sickle cell disease; moreover, fetal hemoglobin functions to replace inadequate hemoglobin A synthesis in β-thalassemia thus serving as an effective therapeutic target. In the perinatal period, fetal hemoglobin is synthesized at high levels followed by a decline to adult levels by one year of age. It is known that naturally occurring mutations in the γ-globin gene promoters and distant cis-acting transcription factors produce persistent fetal hemoglobin synthesis after birth to ameliorate clinical symptoms. Major repressor proteins that silence γ-globin during development have been targeted for gene therapy in β-hemoglobinopathies patients. In parallel effort, several classes of pharmacological agents that induce fetal hemoglobin expression through molecular and cell signaling mechanisms have been identified. Herein, we reviewed the progress made in the discovery of signaling molecules targeted by pharmacologic agents that enhance γ-globin expression and have the potential for future drug development to treat the β-hemoglobinopathies.
Collapse
Affiliation(s)
- Betty S Pace
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA 30912, USA
| | - Li Liu
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75083, USA
| | - Biaoru Li
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA
| | - Levi H Makala
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Pule GD, Mowla S, Novitzky N, Wiysonge CS, Wonkam A. A systematic review of known mechanisms of hydroxyurea-induced fetal hemoglobin for treatment of sickle cell disease. Expert Rev Hematol 2015; 8:669-79. [PMID: 26327494 DOI: 10.1586/17474086.2015.1078235] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AIM To report on molecular mechanisms of fetal hemoglobin (HbF) induction by hydroxyurea (HU) for the treatment of sickle cell disease. STUDY DESIGN Systematic review. RESULTS Studies have provided consistent associations between genomic variations in HbF-promoting loci and variable HbF level in response to HU. Numerous signal transduction pathways have been implicated, through the identification of key genomic variants in BCL11A, HBS1L-MYB, SAR1 or XmnI polymorphism that predispose the response to the treatment, and signal transduction pathways that modulate γ-globin expression (cAMP/cGMP; Giα/c-Jun N-terminal kinase/Jun; methylation and miRNA). Three main molecular pathways have been reported: i) Epigenetic modifications, transcriptional events and signaling pathways involved in HU-mediated response, ii) Signaling pathways involving HU-mediated response and iii) Post-transcriptional pathways (regulation by miRNAs). CONCLUSIONS The complete picture of HU-mediated mechanisms of HbF production in Sickle Cell Disease remains elusive. Research on post-transcriptional mechanisms could lead to therapeutic targets that may minimize alterations to the cellular transcriptome.
Collapse
Affiliation(s)
- Gift D Pule
- a 1 Department of Medicine, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, Republic of South Africa
| | | | | | | | | |
Collapse
|
16
|
Molecular mechanisms underlying synergistic adhesion of sickle red blood cells by hypoxia and low nitric oxide bioavailability. Blood 2014; 123:1917-26. [PMID: 24429338 DOI: 10.1182/blood-2013-06-510180] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The molecular mechanisms by which nitric oxide (NO) bioavailability modulates the clinical expression of sickle cell disease (SCD) remain elusive. We investigated the effect of hypoxia and NO bioavailability on sickle red blood cell (sRBC) adhesion using mice deficient for endothelial NO synthase (eNOS) because their NO metabolite levels are similar to those of SCD mice but without hypoxemia. Whereas sRBC adhesion to endothelial cells in eNOS-deficient mice was synergistically upregulated at the onset of hypoxia, leukocyte adhesion was unaffected. Restoring NO metabolite levels to physiological levels markedly reduced sRBC adhesion to levels seen under normoxia. These results indicate that sRBC adherence to endothelial cells increases in response to hypoxia prior to leukocyte adherence, and that low NO bioavailability synergistically upregulates sRBC adhesion under hypoxia. Although multiple adhesion molecules mediate sRBC adhesion, we found a central role for P-selectin in sRBC adhesion. Hypoxia and low NO bioavailability upregulated P-selectin expression in endothelial cells in an additive manner through p38 kinase pathways. These results demonstrate novel cellular and signaling mechanisms that regulate sRBC adhesion under hypoxia and low NO bioavailability. Importantly, these findings point us toward new molecular targets to inhibit cell adhesion in SCD.
Collapse
|
17
|
Ikuta T, Kuroyanagi Y, Odo N, Liu S. A common signaling pathway is activated in erythroid cells expressing high levels of fetal hemoglobin: a potential role for cAMP-elevating agents in β-globin disorders. J Blood Med 2013; 4:149-59. [PMID: 24353450 PMCID: PMC3862583 DOI: 10.2147/jbm.s54671] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Although erythroid cells prepared from fetal liver, cord blood, or blood from β-thalassemia patients are known to express fetal hemoglobin at high levels, the underlying mechanisms remain elusive. We previously showed that cyclic nucleotides such as cAMP and cGMP induce fetal hemoglobin expression in primary erythroid cells. Here we report that cAMP signaling contributes to high-level fetal hemoglobin expression in erythroid cells prepared from cord blood and β-thalassemia. Methods The status of the cAMP signaling pathway was investigated using primary erythroid cells prepared from cord blood and the mononuclear cells of patients with β-thalassemia; erythroid cells from adult bone marrow mononuclear cells served as the control. Results We found that intracellular cAMP levels were higher in erythroid cells from cord blood and β-thalassemia than from adult bone marrow. Protein kinase A activity levels and cAMP-response element binding protein phosphorylation were higher in erythroid cells from cord blood or β-thalassemia than in adult bone marrow progenitors. Mitogen-activated protein kinase pathways, which play a role in fetal hemoglobin expression, were not consistently activated in cord blood or β-thalassemia erythroid cells. When cAMP signaling was activated in adult erythroid cells, fetal hemoglobin was induced at high levels and associated with reduced expression of BCL11A, a silencer of the β-globin gene. Conclusion These results suggest that activated cAMP signaling may be a common mechanism among erythroid cells with high fetal hemoglobin levels, in part because of downregulation of BCL11A. Activation of the cAMP signaling pathway with cAMP-elevating agents may prove to be an important signaling mechanism to reactivate fetal hemoglobin expression in erythroid cells.
Collapse
Affiliation(s)
- Tohru Ikuta
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Yuichi Kuroyanagi
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Nadine Odo
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Siyang Liu
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
18
|
Vankayala SL, Hargis JC, Woodcock HL. How does catalase release nitric oxide? A computational structure-activity relationship study. J Chem Inf Model 2013; 53:2951-61. [PMID: 24087936 DOI: 10.1021/ci400395c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydroxyurea (HU) is the only FDA approved medication for treating sickle cell disease in adults. The primary mechanism of action is pharmacological elevation of nitric oxide (NO) levels which induces propagation of fetal hemoglobin. HU is known to undergo redox reactions with heme based enzymes like hemoglobin and catalase to produce NO. However, specific details about the HU based NO release remain unknown. Experimental studies indicate that interaction of HU with human catalase compound I produces NO. Presently, we combine flexible receptor-flexible substrate induced fit docking (IFD) with energy decomposition analyses to examine the atomic level details of a possible key step in the clinical conversion of HU to NO. Substrate binding modes of nine HU analogs with catalase compound I were investigated to determine the essential properties necessary for effective NO release. Three major binding orientations were found that provide insight into the possible reaction mechanisms for producing NO. Further results show that anion/radical intermediates produced as part of these mechanisms would be stabilized by hydrogen bonding interactions from distal residues His75, Asn148, Gln168, and oxoferryl-heme. These details will ideally contribute to both a clearer mechanistic picture and provide insights for future structure based drug design efforts.
Collapse
Affiliation(s)
- Sai Lakshmana Vankayala
- Department of Chemistry, University of South Florida , 4202 E. Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | | | | |
Collapse
|
19
|
Banan M, Esmaeilzadeh-Gharehdaghi E, Nezami M, Deilami Z, Farashi S, Philipsen S, Esteghamat F, Pourfarzad F, Ali Imam AM, Najmabadi H. cAMP response element-binding protein 1 is required for hydroxyurea-mediated induction of γ-globin expression in K562 cells. Clin Exp Pharmacol Physiol 2013; 39:510-7. [PMID: 22469229 DOI: 10.1111/j.1440-1681.2012.05702.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
1. Hydroxyurea (HU) is a drug used for the treatment of haemoglobinopathies. Hydroxyurea functions by upregulating γ-globin transcription and fetal haemoglobin (HbF) production in erythroid cells. The K562 erythroleukaemia cell line is widely used as a model system in which to study the mechanism of γ-globin induction by HU. However, the transcription factors required for the upregulation of γ-globin expression by HU in K562 cells have not been identified. Similarities between the HU and sodium butyrate (SB) pathways suggest cAMP response element-binding protein (CREB) 1 as a potential candidate. Thus, the aim of the present study was to investigate the possible role of CREB1 in the HU pathway. 2. Experiments were performed using transient and stable RNA interference (RNAi) to show that CREB1 is necessary for HU-mediated induction of γ-globin expression and haemoglobin production in K562 cells. 3. Furthermore, western blot analyses demonstrated that CREB1 becomes phosphorylated in a dose-dependent manner after HU (100-400 µmol/L) treatment of K562 cells for 72 h. 4. We also investigated role of a Gγ promoter CREB1 response element (G-CRE) in this pathway. Quantitative amplification refractory mutation system-polymerase chain reaction experiments were performed to demonstrate that HU induces the expression of both Gγ and Aγ in this cell line. In addition, electrophoretic mobility shift assays were used to show that levels of CREB1 complexes binding to the G-CRE site are increased following HU treatment and are decreased in CREB1-knockdown cells. 5. The results suggest that CREB1 is necessary for γ-globin induction by HU in K562 cells, a role that may be mediated, in part, through the G-CRE element.
Collapse
Affiliation(s)
- Mehdi Banan
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Čokić VP, Smith RD, Biancotto A, Noguchi CT, Puri RK, Schechter AN. Globin gene expression in correlation with G protein-related genes during erythroid differentiation. BMC Genomics 2013; 14:116. [PMID: 23425329 PMCID: PMC3602204 DOI: 10.1186/1471-2164-14-116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 02/11/2013] [Indexed: 12/22/2022] Open
Abstract
Background The guanine nucleotide binding protein (G protein)-coupled receptors (GPCRs) regulate cell growth, proliferation and differentiation. G proteins are also implicated in erythroid differentiation, and some of them are expressed principally in hematopoietic cells. GPCRs-linked NO/cGMP and p38 MAPK signaling pathways already demonstrated potency for globin gene stimulation. By analyzing erythroid progenitors, derived from hematopoietic cells through in vitro ontogeny, our study intends to determine early markers and signaling pathways of globin gene regulation and their relation to GPCR expression. Results Human hematopoietic CD34+ progenitors are isolated from fetal liver (FL), cord blood (CB), adult bone marrow (BM), peripheral blood (PB) and G-CSF stimulated mobilized PB (mPB), and then differentiated in vitro into erythroid progenitors. We find that growth capacity is most abundant in FL- and CB-derived erythroid cells. The erythroid progenitor cells are sorted as 100% CD71+, but we did not find statistical significance in the variations of CD34, CD36 and GlyA antigens and that confirms similarity in maturation of studied ontogenic periods. During ontogeny, beta-globin gene expression reaches maximum levels in cells of adult blood origin (176 fmol/μg), while gamma-globin gene expression is consistently up-regulated in CB-derived cells (60 fmol/μg). During gamma-globin induction by hydroxycarbamide, we identify stimulated GPCRs (PTGDR, PTGER1) and GPCRs-coupled genes known to be activated via the cAMP/PKA (ADIPOQ), MAPK pathway (JUN) and NO/cGMP (PRPF18) signaling pathways. During ontogeny, GPR45 and ARRDC1 genes have the most prominent expression in FL-derived erythroid progenitor cells, GNL3 and GRP65 genes in CB-derived cells (high gamma-globin gene expression), GPR110 and GNG10 in BM-derived cells, GPR89C and GPR172A in PB-derived cells, and GPR44 and GNAQ genes in mPB-derived cells (high beta-globin gene expression). Conclusions These results demonstrate the concomitant activity of GPCR-coupled genes and related signaling pathways during erythropoietic stimulation of globin genes. In accordance with previous reports, the stimulation of GPCRs supports the postulated connection between cAMP/PKA and NO/cGMP pathways in activation of γ-globin expression, via JUN and p38 MAPK signaling.
Collapse
Affiliation(s)
- Vladan P Čokić
- Laboratory of Experimental Hematology, Institute for Medical Research, University of Belgrade, Dr, Subotica 4, 11129, Belgrade, Serbia.
| | | | | | | | | | | |
Collapse
|
21
|
Rahim F, Allahmoradi H, Salari F, Shahjahani M, Fard AD, Hosseini SA, Mousakhani H. Evaluation of Signaling Pathways Involved in γ-Globin Gene Induction Using Fetal Hemoglobin Inducer Drugs. Int J Hematol Oncol Stem Cell Res 2013; 7:41-6. [PMID: 24505534 PMCID: PMC3913148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 06/15/2013] [Indexed: 11/08/2022] Open
Abstract
Potent induction of fetal hemoglobin (HbF) production results in alleviating the complications of β-thalassemia and sickle cell disease (SCD). HbF inducer agents can trigger several molecular signaling pathways critical for erythropoiesis. Janus kinase/Signal transducer and activator of transcription (JAK/STAT), mitogen activated protein kinas (MAPK) and Phosphoinositide 3-kinase (PI3K) are considered as main signaling pathways, which may play a significant role in HbF induction. All these signaling pathways are triggered by erythropoietin (EPO) as the main growth factor inducing erythroid differentiation, when it binds to its cell surface receptor, erythropoietin receptor (EPO-R) HbF inducer agents have been shown to upregulate HbF production level by triggering certain signaling pathways. As a result, understanding the pivotal signaling pathways influencing HbF induction leads to effective upregulation of HbF. In this mini review article, we try to consider the correlation between HbF inducer agents and their molecular mechanisms of γ-globin upregulation. Several studies suggest that activating P38 MAPK, RAS and STAT5 signaling pathways result in efficient HbF induction. Nevertheless, the role of other erythroid signaling pathways in HbF induction seems to be indispensible and should be emphasized.
Collapse
Affiliation(s)
- Fakher Rahim
- Toxicology Research Center, Ahvaz University of Medical Sciences, Ahvaz, Iran
| | - Hossein Allahmoradi
- General Practitioner, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Salari
- Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Shahjahani
- Department of Hematology and Blood Banking, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Dehghani Fard
- Sarem Cell Research Center- SCRC, Sarem Women's Hospital, Tehran, Iran
| | - Seyed Ahmad Hosseini
- Department of nutrition, Allied Health Sciences School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Mousakhani
- Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
22
|
Hydroxyurea and a cGMP-amplifying agent have immediate benefits on acute vaso-occlusive events in sickle cell disease mice. Blood 2012; 120:2879-88. [PMID: 22833547 DOI: 10.1182/blood-2012-02-409524] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Inhibition of leukocyte adhesion to the vascular endothelium represents a novel and important approach for decreasing sickle cell disease (SCD) vaso-occlusion. Using a humanized SCD-mouse-model of tumor necrosis factor-α-induced acute vaso-occlusion, we herein present data demonstrating that short-term administration of either hydroxyurea or the phosphodiesterase 9 (PDE9) inhibitor, BAY73-6691, significantly altered leukocyte recruitment to the microvasculature. Notably, the administration of both agents led to marked improvements in leukocyte rolling and adhesion and decreased heterotypic red blood cell-leukocyte interactions, coupled with prolonged animal survival. Mechanistically, these rheologic benefits were associated with decreased endothelial adhesion molecule expression, as well as diminished leukocyte Mac-1-integrin activation and cyclic guanosine monophosphate (cGMP)-signaling, leading to reduced leukocyte recruitment. Our findings indicate that hydroxyurea has immediate beneficial effects on the microvasculature in acute sickle-cell crises that are independent of the drug's fetal hemoglobin-elevating properties and probably involve the formation of intravascular nitric oxide. In addition, inhibition of PDE9, an enzyme highly expressed in hematopoietic cells, amplified the cGMP-elevating effects of hydroxyurea and may represent a promising and more tissue-specific adjuvant therapy for this disease.
Collapse
|
23
|
Flanagan JM, Steward S, Howard TA, Mortier NA, Kimble AC, Aygun B, Hankins JS, Neale GA, Ware RE. Hydroxycarbamide alters erythroid gene expression in children with sickle cell anaemia. Br J Haematol 2012; 157:240-8. [PMID: 22360576 DOI: 10.1111/j.1365-2141.2012.09061.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 12/12/2011] [Indexed: 01/16/2023]
Abstract
Sickle cell anaemia (SCA) is a severe debilitating haematological disorder associated with a high degree of morbidity and mortality. The level of fetal haemoglobin (HbF) is well-recognized as a critical laboratory parameter: lower HbF is associated with a higher risk of vaso-occlusive complications, organ damage, and early death. Hydroxycarbamide treatment can induce HbF, improve laboratory parameters, and ameliorate clinical complications of SCA but its mechanisms of action remain incompletely defined and the HbF response is highly variable. To identify pathways of hydroxycarbamide activity, we performed microarray expression analyses of early reticulocyte RNA obtained from children with SCA enrolled in the HydroxyUrea Study of Long-term Effects (NCT00305175) and examined the effects of hydroxycarbamide exposure in vivo. Hydroxycarbamide affected a large number of erythroid genes, with significant decreases in the expression of genes involved in translation, ribosome assembly and chromosome organization, presumably reflecting the daily cytotoxic pulses of hydroxycarbamide. Hydroxycarbamide also affected expression of numerous genes associated with HbF including BCL11A, a key regulator of baseline HbF levels. Together, these data indicate that hydroxycarbamide treatment for SCA leads to substantial changes in erythroid gene expression, including BCL11A and other potential signalling pathways associated with HbF induction.
Collapse
Affiliation(s)
- Jonathan M Flanagan
- International Hematology Center of Excellence, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ikuta T, Adekile AD, Gutsaeva DR, Parkerson JB, Yerigenahally SD, Clair B, Kutlar A, Odo N, Head CA. The proinflammatory cytokine GM-CSF downregulates fetal hemoglobin expression by attenuating the cAMP-dependent pathway in sickle cell disease. Blood Cells Mol Dis 2011; 47:235-42. [PMID: 21945571 PMCID: PMC3223356 DOI: 10.1016/j.bcmd.2011.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 08/10/2011] [Accepted: 08/20/2011] [Indexed: 02/02/2023]
Abstract
Although reduction in leukocyte counts following hydroxyurea therapy in sickle cell disease (SCD) predicts fetal hemoglobin (HbF) response, the underlying mechanism remains unknown. We previously reported that leukocyte counts are regulated by granulocyte-macrophage colony-stimulating factor (GM-CSF) in SCD patients. Here we examined the roles of GM-CSF in the regulation of HbF expression in SCD. Upon the analysis of retrospective data in 372 patients, HbF levels were inversely correlated with leukocyte counts and GM-CSF levels in SCD patients without hydroxyurea therapy, while HbF increments after hydroxyurea therapy correlated with a reduction in leukocyte counts, suggesting a negative effect of GM-CSF on HbF expression. Consistently, in vitro studies using primary erythroblasts showed that the addition of GM-CSF to erythroid cells decreased HbF expression. We next examined the intracellular signaling pathway through which GM-CSF reduced HbF expression. Treatment of erythroid cells with GM-CSF resulted in the reduction of intracellular cAMP levels and abrogated phosphorylation of cAMP response-element-binding-protein, suggesting attenuation of the cAMP-dependent pathway, while the phosphorylation levels of mitogen-activated protein kinases were not affected. This is compatible with our studies showing a role for the cAMP-dependent pathway in HbF expression. Together, these results demonstrate that GM-CSF plays a role in regulating both leukocyte count and HbF expression in SCD. Reduction in GM-CSF levels upon hydroxyurea therapy may be critical for efficient HbF induction. The results showing the involvement of GM-CSF in HbF expression may suggest possible mechanisms for hydroxyurea resistance in SCD.
Collapse
Affiliation(s)
- Tohru Ikuta
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Georgia Health Sciences University, Augusta, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Sickle cell anemia (SCA) is a well characterized severe hematological disorder with substantial morbidity and early mortality. Hydroxyurea is a potent inducer of fetal hemoglobin, and evidence over the past 25 years has documented its laboratory and clinical efficacy for both adults and children with SCA. RECENT FINDINGS The phase III study of hydroxyurea in infants (BABY HUG) has just been completed and preliminary results indicate equivocal benefits for organ protection during the 2-year treatment period, but significant benefits for pain, acute chest syndrome, hospitalizations, and transfusions. Three new reports document the benefits of hydroxyurea on reducing mortality in SCA: two adult trials (LaSHS and MSH) and one pediatric study (Brazilian cohort). Recent results from the HUSTLE protocol suggest minimal genotoxicity or carcinogenicity with long-term hydroxyurea exposure. SUMMARY The potential utility of hydroxyurea for all patients with SCA is clear and indisputable. With decades of accumulated evidence and documented efficacy with an acceptable long-term safety profile, it is time to consider hydroxyurea treatment the standard of care for all young patients with SCA. Exporting our knowledge and experience with hydroxyurea to developing nations with large medical burdens from SCA can help relieve global suffering from this condition.
Collapse
Affiliation(s)
- Patrick T McGann
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| | | |
Collapse
|
26
|
Correia MA, Sinclair PR, De Matteis F. Cytochrome P450 regulation: the interplay between its heme and apoprotein moieties in synthesis, assembly, repair, and disposal. Drug Metab Rev 2011; 43:1-26. [PMID: 20860521 PMCID: PMC3034403 DOI: 10.3109/03602532.2010.515222] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Heme is vital to our aerobic universe. Heme cellular content is finely tuned through an exquisite control of synthesis and degradation. Heme deficiency is deleterious to cells, whereas excess heme is toxic. Most of the cellular heme serves as the prosthetic moiety of functionally diverse hemoproteins, including cytochromes P450 (P450s). In the liver, P450s are its major consumers, with >50% of hepatic heme committed to their synthesis. Prosthetic heme is the sine qua non of P450 catalytic biotransformation of both endo- and xenobiotics. This well-recognized functional role notwithstanding, heme also regulates P450 protein synthesis, assembly, repair, and disposal. These less well-appreciated aspects are reviewed herein.
Collapse
Affiliation(s)
- Maria Almira Correia
- Department of Cellular and Molecular Pharmacology, The Liver Center, University of California, San Francisco, 94158, USA.
| | | | | |
Collapse
|
27
|
Atweh G, Fathallah H. Pharmacologic induction of fetal hemoglobin production. Hematol Oncol Clin North Am 2010; 24:1131-44. [PMID: 21075284 DOI: 10.1016/j.hoc.2010.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Reactivation of fetal hemoglobin (HbF) expression is an important therapeutic option in adult patients with hemoglobin disorders. The understanding of the developmental regulation of γ-globin gene expression was followed by the identification of a number of chemical compounds that can reactivate HbF synthesis in vitro and in vivo in patients with hemoglobin disorders. These HbF inducers can be grouped in several classes based on their mechanisms of action. This article focuses on pharmacologic agents that were tested in humans and discusses current knowledge about the mechanisms by which they induce HbF.
Collapse
Affiliation(s)
- George Atweh
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0562, USA
| | | |
Collapse
|
28
|
Cho CS, Kato GJ, Yang SH, Bae SW, Lee JS, Gladwin MT, Rhee SG. Hydroxyurea-induced expression of glutathione peroxidase 1 in red blood cells of individuals with sickle cell anemia. Antioxid Redox Signal 2010; 13:1-11. [PMID: 19951064 PMCID: PMC2935334 DOI: 10.1089/ars.2009.2978] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Chronic redox imbalance in erythrocytes of individuals with sickle cell disease (SCD) contributes to oxidative stress and likely underlies common etiologies of hemolysis. We measured the amounts of six antioxidant enzymes-SOD1, catalase, glutathione peroxidase 1 (GPx1), as well as peroxiredoxins (Prxs) I, II, and VI-in red blood cells (RBCs) of SCD patients and control subjects. The amounts of SOD1 and Prx VI were reduced by about 17% and 20%, respectively, in SCD RBCs compared with control cells. The amounts of Prx II and GPx1 did not differ between SCD and normal RBCs. However, about 18% of Prx II was inactivated in SCD RBCs as a result of oxidation to sulfinic Prx II, whereas inactive Prx II was virtually undetectable in control cells. Furthermore, GPx1 activity was reduced by about 33% in SCD RBCs, and the loss of activity was correlated with hemolysis in SCD patients. RBCs from SCD patients taking hydroxyurea demonstrated 90% higher GPx1 activity than did those from untreated SCD patients, with no differences seen for the other catalytic antioxidants. Hydroxyurea induced GPx1 expression in multiple cultured cell lines in a manner dependent on both p53 and NO-cGMP signaling pathways. GPx1 expression represents a previously unrecognized potential benefit of hydroxyurea treatment in SCD patients.
Collapse
Affiliation(s)
- Chun-Seok Cho
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seodaemun-gu, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
Cokić VP, Beleslin-Cokić BB, Smith RD, Economou AP, Wahl LM, Noguchi CT, Schechter AN. Stimulated stromal cells induce gamma-globin gene expression in erythroid cells via nitric oxide production. Exp Hematol 2009; 37:1230-7. [PMID: 19576950 PMCID: PMC2748161 DOI: 10.1016/j.exphem.2009.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 06/15/2009] [Accepted: 06/26/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We have previously shown that nitric oxide (NO) is involved in the hydroxyurea-induced increase of gamma-globin gene expression in cultured human erythroid progenitor cells and that hydroxyurea increases NO production in endothelial cells via endothelial NO synthase (NOS). We have now expanded those studies to demonstrate that stimulation of gamma-globin gene expression is also mediated by NOS induction in stromal cells within the bone marrow microenvironment. MATERIALS AND METHODS Using NO analyzer, we measured NO production in endothelial and macrophage cell cultures. In coculture studies of erythroid and stromal cells, we measured globin gene expression during stimulation by NO inducers. RESULTS Hydroxyurea (30-100 microM) induced NOS-dependent production of NO in human macrophages (up to 1.2 microM). Coculture studies of human macrophages with erythroid progenitor cells also resulted in induction of gamma-globin mRNA expression (up to threefold) in the presence of hydroxyurea. NOS-dependent stimulation of NO by lipopolysaccharide (up to 0.6 microM) has been observed in human macrophages. We found that lipopolysaccharide and interferon-gamma together increased gamma-globin gene expression (up to twofold) in human macrophage/erythroid cell cocultures. Coculture of human bone marrow endothelial cells with erythroid progenitor cells also induced gamma-globin mRNA expression (2.4-fold) in the presence of hydroxyurea (40 microM). CONCLUSION These results demonstrate an arrangement by which NO and fetal hemoglobin inducers may stimulate globin genes in erythroid cells via the common paracrine effect of bone marrow stromal cells.
Collapse
Affiliation(s)
- Vladan P Cokić
- Laboratory of Experimental Hematology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia.
| | | | | | | | | | | | | |
Collapse
|
30
|
Cytokine-mediated increases in fetal hemoglobin are associated with globin gene histone modification and transcription factor reprogramming. Blood 2009; 114:2299-306. [PMID: 19597182 DOI: 10.1182/blood-2009-05-219386] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Therapeutic regulation of globin genes is a primary goal of translational research aimed toward hemoglobinopathies. Signal transduction was used to identify chromatin modifications and transcription factor expression patterns that are associated with globin gene regulation. Histone modification and transcriptome profiling were performed using adult primary CD34(+) cells cultured with cytokine combinations that produced low versus high levels of gamma-globin mRNA and fetal hemoglobin (HbF). Embryonic, fetal, and adult globin transcript and protein expression patterns were determined for comparison. Chromatin immunoprecipitation assays revealed RNA polymerase II occupancy and histone tail modifications consistent with transcriptional activation only in the high-HbF culture condition. Transcriptome profiling studies demonstrated reproducible changes in expression of nuclear transcription factors associated with high HbF. Among the 13 genes that demonstrated differential transcript levels, 8 demonstrated nuclear protein expression levels that were significantly changed by cytokine signal transduction. Five of the 8 genes are recognized regulators of erythropoiesis or globin genes (MAFF, ID2, HHEX, SOX6, and EGR1). Thus, cytokine-mediated signal transduction in adult erythroid cells causes significant changes in the pattern of globin gene and protein expression that are associated with distinct histone modifications as well as nuclear reprogramming of erythroid transcription factors.
Collapse
|
31
|
Conran N, Costa FF. Hemoglobin disorders and endothelial cell interactions. Clin Biochem 2009; 42:1824-38. [PMID: 19580799 DOI: 10.1016/j.clinbiochem.2009.06.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Accepted: 06/20/2009] [Indexed: 11/15/2022]
Abstract
Endothelial damage and inflammation make a significant contribution to the pathophysiology of sickle cell disease (SCD) and the beta-thalassemia syndromes. Endothelial dysfunction and ensuing vasculopathy are implicated in pulmonary hypertension in the hemoglobinopathies and endothelial activation and endothelial-blood cell adhesion, accompanied by inflammatory processes and oxidative stress, are imperative to the vaso-occlusive process in SCD. Herein, we discuss the role that the endothelium plays in all of these processes and the effect that genetic modifiers and hydroxyurea therapy may have upon endothelial interactions. Therapies targeting the endothelium and endothelial interactions may represent a promising approach for treating these diseases.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology and Hemotherapy Centre, School of Medical Sciences, University of Campinas - UNICAMP, Brazil.
| | | |
Collapse
|
32
|
Perrine SP, Mankidy R, Boosalis MS, Bieker JJ, Faller DV. Erythroid Kruppel-like factor (EKLF) is recruited to the gamma-globin gene promoter as a co-activator and is required for gamma-globin gene induction by short-chain fatty acid derivatives. Eur J Haematol 2009; 82:466-76. [PMID: 19220418 PMCID: PMC3232177 DOI: 10.1111/j.1600-0609.2009.01234.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The erythroid Kruppel-like factor (EKLF) is an essential transcription factor for beta-type globin gene switching, and specifically activates transcription of the adult beta-globin gene promoter. We sought to determine if EKLF is also required for activation of the gamma-globin gene by short-chain fatty acid (SCFA) derivatives, which are now entering clinical trials. METHODS The functional and physical interaction of EKLF and co-regulatory molecules with the endogenous human globin gene promoters was studied in primary human erythroid progenitors and cell lines, using chromatin immunoprecipitation (ChIP) assays and genetic manipulation of the levels of EKLF and co-regulators. RESULTS AND CONCLUSIONS Knockdown of EKLF prevents SCFA-induced expression of the gamma-globin promoter in a stably expressed microLCRbeta(pr)R(luc) (A)gamma(pr)F(luc) cassette, and prevents induction of the endogenous gamma-globin gene in primary human erythroid progenitors. EKLF is actively recruited to endogenous gamma-globin gene promoters after exposure of primary human erythroid progenitors, and murine hematopoietic cell lines, to SCFA derivatives. The core ATPase BRG1 subunit of the human SWI/WNF complex, a ubiquitous multimeric complex that regulates gene expression by remodeling nucleosomal structure, is also required for gamma-globin gene induction by SCFA derivatives. BRG1 is actively recruited to the endogenous gamma-globin promoter of primary human erythroid progenitors by exposure to SCFA derivatives, and this recruitment is dependent upon the presence of EKLF. These findings demonstrate that EKLF, and the co-activator BRG1, previously demonstrated to be required for definitive or adult erythropoietic patterns of globin gene expression, are co-opted by SCFA derivatives to activate the fetal globin genes.
Collapse
Affiliation(s)
- Susan P. Perrine
- Cancer Center, Boston University School of Medicine, Boston, MA
- Hemoglobinopathy-Thalassemia Research Unit, Departments of Medicine, Pediatrics, Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA
- Phoenicia Biosciences, Inc., Newton, MA
| | - Rishikesh Mankidy
- Cancer Center, Boston University School of Medicine, Boston, MA
- Hemoglobinopathy-Thalassemia Research Unit, Departments of Medicine, Pediatrics, Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA
| | - Michael S. Boosalis
- Cancer Center, Boston University School of Medicine, Boston, MA
- Hemoglobinopathy-Thalassemia Research Unit, Departments of Medicine, Pediatrics, Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA
| | | | | |
Collapse
|
33
|
Short-chain fatty acid-mediated effects on erythropoiesis in primary definitive erythroid cells. Blood 2009; 113:6440-8. [PMID: 19380871 DOI: 10.1182/blood-2008-09-171728] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Short-chain fatty acids (SCFAs; butyrate and propionate) up-regulate embryonic/fetal globin gene expression through unclear mechanisms. In a murine model of definitive erythropoiesis, SCFAs increased embryonic beta-type globin gene expression in primary erythroid fetal liver cells (eFLCs) after 72 hours in culture, from 1.7% (+/- 1.2%) of total beta-globin gene expression at day 0 to 4.9% (+/- 2.2%) in propionate and 5.4% (+/- 3.4%) in butyrate; this effect was greater in butyrate plus insulin/erythropoietin (BIE), at 19.5% (+/- 8.3%) compared with 0.1% (+/- 0.1%) in ins/EPO alone (P < .05). Fetal gamma-globin gene expression was increased in human transgene-containing eFLCs, to 35.9% (+/- 7.0%) in BIE compared with 4.4% (+/- 4.2%) in ins/EPO only (P < .05). Embryonic globin gene expression was detectable in 11 of 15 single eFLCs treated with BIE, but in0 of 15 ins/EPO-only treated cells. Butyrate-treated [65.5% (+/- 9.9%)] and 77.5% (+/- 4.0%) propionate-treated eFLCs were highly differentiated in culture, compared with 21.5% (+/- 3.5%) in ins/EPO (P < .005). Importantly, signaling intermediaries, previously implicated in induced embryonic/fetal globin gene expression (STAT5, p42/44, and p38), were not differentially activated by SCFAs in eFLCs; but increased bulk histone (H3) acetylation was seen in SCFA-treated eFLCs. SCFAs induce embryonic globin gene expression in eFLCS, which are a useful short-term and physiologic primary cell model of embryonic/fetal globin gene induction during definitive erythropoiesis.
Collapse
|
34
|
Little JA, Hauser KP, Martyr SE, Harris A, Maric I, Morris CR, Suh JH, Taylor J, Castro O, Machado R, Kato G, Gladwin MT. Hematologic, biochemical, and cardiopulmonary effects of L-arginine supplementation or phosphodiesterase 5 inhibition in patients with sickle cell disease who are on hydroxyurea therapy. Eur J Haematol 2009; 82:315-21. [PMID: 19215288 PMCID: PMC2775051 DOI: 10.1111/j.1600-0609.2009.01210.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Fetal hemoglobin (HbF) induction involves NO-cGMP signaling pathways. L-arginine, an NO precursor, and the phosphodiesterase (PDE) 5 inhibitor sildenafil, which potentiates cGMP, were studied in adults with sickle cell disease (SCD) who were stably on HU. METHODS Twenty four courses of L-arginine (0.1-0.2 g/kg divided TID) or sildenafil (25-100 mg TID), assigned based on gender due to concerns about sildenafil-related priapism, were successfully completed. Biochemical assays, pulmonary pressures, and cardiopulmonary exercise capacity are reported from patients in whom serial values are available. Hematologic responses are reported in 14 subjects with HbSS who had stable baseline HbF levels. RESULTS L-arginine increased plasma arginine and ornithine, but not citrulline, suggesting diversion by plasma arginase from NO, and citrulline, generation. Glutathione increased only in patients on L-arginine. Sildenafil increased plasma cGMP and citrulline, but not other amino acids. Pulmonary pressures and 6-min walk distances improved only in patients on sildenafil. In subjects with stable baseline HbF levels, HbF levels changed little from a normalized baseline on l-arginine, decreasing by 2.9 +/- 16.1%, n = 6; P = n.s., but increased on sildenafil, by 7.5 +/- 11.7%, n = 8, P < 0.05. Absolute reticulocyte counts initially decreased in patients on sildenafil. CONCLUSIONS L-arginine, at doses that increase plasma arginine levels, altered redox potential in red cells. The lack of clinically detectable efficacy of L-arginine may be due to increased arginine metabolism in SCD patients. In vivo augmentation of the cyclic nucleotide pathway by PDE inhibition may induce HbF slightly, but strikingly improves hemodynamic and functional status in SCD.
Collapse
Affiliation(s)
- Jane A Little
- Pulmonary and Vascular Medicine Branch, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892-1476, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Moreira LS, de Andrade TG, Albuquerque DM, Cunha AF, Fattori A, Saad STO, Costa FF. IDENTIFICATION OF DIFFERENTIALLY EXPRESSED GENES INDUCED BY HYDROXYUREA IN RETICULOCYTES FROM SICKLE CELL ANAEMIA PATIENTS. Clin Exp Pharmacol Physiol 2008; 35:651-5. [DOI: 10.1111/j.1440-1681.2007.04861.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
36
|
Abstract
To explore the functional significance of cGMP-dependent protein kinase type I (cGKI) in the regulation of erythrocyte survival, gene-targeted mice lacking cGKI were compared with their control littermates. By the age of 10 weeks, cGKI-deficient mice exhibited pronounced anemia and splenomegaly. Compared with control mice, the cGKI mutants had significantly lower red blood cell count, packed cell volume, and hemoglobin concentration. Anemia was associated with a higher reticulocyte number and an increase of plasma erythropoietin concentration. The spleens of cGKI mutant mice were massively enlarged and contained a higher fraction of Ter119(+) erythroid cells, whereas the relative proportion of leukocyte subpopulations was not changed. The Ter119(+) cGKI-deficient splenocytes showed a marked increase in annexin V binding, pointing to phosphatidylserine (PS) exposure at the outer membrane leaflet, a hallmark of suicidal erythrocyte death or eryptosis. Compared with control erythrocytes, cGKI-deficient erythrocytes exhibited in vitro a higher cytosolic Ca(2+) concentration, a known trigger of eryptosis, and showed increased PS exposure, which was paralleled by a faster clearance in vivo. Together, these results identify a role of cGKI as mediator of erythrocyte survival and extend the emerging concept that cGMP/cGKI signaling has an antiapoptotic/prosurvival function in a number of cell types in vivo.
Collapse
|
37
|
Kumkhaek C, Taylor JG, Zhu J, Hoppe C, Kato GJ, Rodgers GP. Fetal haemoglobin response to hydroxycarbamide treatment and sar1a promoter polymorphisms in sickle cell anaemia. Br J Haematol 2008; 141:254-9. [PMID: 18318767 PMCID: PMC2344124 DOI: 10.1111/j.1365-2141.2008.07045.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 12/05/2007] [Indexed: 11/30/2022]
Abstract
The hydroxycarbamide (HC)-inducible small guanosine triphosphate (GTP)-binding protein, secretion-associated and RAS-related (SAR) protein has recently been shown to play a pivotal role in HBG induction and erythroid maturation by causing cell apoptosis and G1/S-phase arrest. Our preliminary analysis indicated that HC inducibility is transcriptionally regulated by elements within the SAR1A promoter. This study aimed to assess whether polymorphisms in the SAR1A promoter are associated with differences Hb F levels or HC therapeutic responses among sickle cell disease (SCD) patients. We studied 386 individuals with SCD comprised of 269 adults treated with or without HC and 117 newborns with SCD identified from a newborn screening program. Three previously unknown single nucleotide polymorphisms (SNPs) in the upstream 5'UTR (-809 C>T, -502 G>T and -385 C>A) were significantly associated with the fetal haemoglobin (HbF) response in Hb SS patients treated with HC (P < 0.05). In addition, four SNPs (rs2310991, -809 C>T, -385 C>A and rs4282891) were significantly associated with the change in absolute HbF after 2 years of treatment with HC. These data suggest that variation within SAR1A regulatory elements might contribute to inter-individual differences in regulation of HbF expression and patient responses to HC in SCD.
Collapse
Affiliation(s)
- Chutima Kumkhaek
- Molecular and Clinical Hematology Branch, NIDDK, NIH, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
38
|
Aslan M, Freeman BA. Redox-dependent impairment of vascular function in sickle cell disease. Free Radic Biol Med 2007; 43:1469-83. [PMID: 17964418 PMCID: PMC2139908 DOI: 10.1016/j.freeradbiomed.2007.08.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 08/23/2007] [Accepted: 08/23/2007] [Indexed: 01/04/2023]
Abstract
The vascular pathophysiology of sickle cell disease (SCD) is influenced by many factors, including adhesiveness of red and white blood cells to endothelium, increased coagulation, and homeostatic perturbation. The vascular endothelium is central to disease pathogenesis because it displays adhesion molecules for blood cells, balances procoagulant and anticoagulant properties of the vessel wall, and regulates vascular homeostasis by synthesizing vasoconstricting and vasodilating substances. The occurrence of intermittent vascular occlusion in SCD leads to reperfusion injury associated with granulocyte accumulation and enhanced production of reactive oxygen species. The participation of nitric oxide (NO) in oxidative reactions causes a reduction in NO bioavailability and contributes to vascular dysfunction in SCD. Therapeutic strategies designed to counteract endothelial, inflammatory, and oxidative abnormalities may reduce the frequency of hospitalization and blood transfusion, the incidence of pain, and the occurrence of acute chest syndrome and pulmonary hypertension in patients with SCD.
Collapse
Affiliation(s)
- Mutay Aslan
- Department of Biochemistry, Akdeniz University School of Medicine, 07070 Antalya, Turkey.
| | | |
Collapse
|
39
|
Abstract
Hydroxyurea, a drug widely used for treating myeloproliferative diseases, has also been approved for the treatment of sickle cell disease by raising fetal hemoglobin (HbF). We have shown that nitric oxide (NO) and the soluble guanylyl cyclase (sGC) pathways are involved in hydroxyurea induction of HbF levels in erythroid progenitor cells (EPCs). We demonstrate now that during erythroid differentiation, endothelial NO synthase mRNA and protein levels decline steadily, as does the production of NO derivatives and cyclic adenosine monophosphate (cAMP) levels, but guanosine 3',5'-cyclic monophosphate (cGMP) levels are stable. Hydroxyurea increased intracellular cGMP levels and cAMP levels in EPCs. The NO donor, DEANONOate, induced much higher cGMP levels, but reduced cAMP levels. Hydroxyurea (1 mM) induced production of approximately 45 pM cGMP/minute/ng of purified sGC, similar to induction by 1 muM DEANONOate. We found that hydroxyurea and ProliNONOate produced iron-nitrosyl derivatives of sGC. Thus, we confirm that hydroxyurea can directly interact with the deoxy-heme of sGC, presumably by a free-radical nitroxide pathway, and activate cGMP production. These data add to an expanding appreciation of the role of hydroxyurea as an inducer of the NO/cGMP pathway in EPCs. These mechanisms may also be involved in the cytostatic effects of hydroxyurea, as well as the induction of HbF.
Collapse
|
40
|
Neither DNA hypomethylation nor changes in the kinetics of erythroid differentiation explain 5-azacytidine's ability to induce human fetal hemoglobin. Blood 2007; 111:411-20. [PMID: 17916742 DOI: 10.1182/blood-2007-06-093948] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
5-azacytidine (5-Aza) is a potent inducer of fetal hemoglobin (HbF) in people with beta-thalassemia and sickle cell disease. Two models have been proposed to explain this activity. The first is based on the drug's ability to inhibit global DNA methylation, including the fetal globin genes, resulting in their activation. The second is based on 5-Aza's cytotoxicity and observations that HbF production is enhanced during marrow recovery. We tested these models using human primary cells in an in vitro erythroid differentiation system. We found that doses of 5-Aza that produce near maximal induction of gamma-globin mRNA and HbF do not alter cell growth, differentiation kinetics, or cell cycle, but do cause a localized demethylation of the gamma promoter. However, when we reduced gamma promoter methylation to levels equivalent to those seen with 5-Aza or to the lower levels seen in primary fetal erythroid cells using DNMT1 siRNA and shRNA, we observed no induction of gamma-globin mRNA or HbF. These results suggest that 5-Aza induction of HbF is not the result of global DNA demethylation or of changes in differentiation kinetics, but involves an alternative, previously unrecognized mechanism. Other results suggest that posttranscriptional regulation plays an important role in the 5-Aza response.
Collapse
|
41
|
Bailey L, Kuroyanagi Y, Franco-Penteado CF, Conran N, Costa FF, Ausenda S, Cappellini MD, Ikuta T. Expression of the gamma-globin gene is sustained by the cAMP-dependent pathway in beta-thalassaemia. Br J Haematol 2007; 138:382-95. [PMID: 17614826 DOI: 10.1111/j.1365-2141.2007.06673.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The present study found that the cyclic adenosine monophosphate (cAMP)-dependent pathway efficiently induced gamma-globin expression in adult erythroblasts, and this pathway plays a role in gamma-globin gene (HBG) expression in beta-thalassaemia. Expression of HBG mRNA increased to about 46% of non-HBA mRNA in adult erythroblasts treated with forskolin, while a cyclic guanosine monophosphate (cGMP) analogue induced HBG mRNA to levels <20% of non-HBA mRNA. In patients with beta-thalassaemia intermedia, cAMP levels were elevated in both red blood cells and nucleated erythroblasts but no consistent elevation was found with cGMP levels. The transcription factor cAMP response element binding protein (CREB) was phosphorylated in nucleated erythroblasts and its phosphorylation levels correlated with HBG mRNA levels of the patients. Other signalling molecules, such as mitogen-activated protein kinases and signal transducers and activators of transcription proteins, were phosphorylated at variable levels and showed no correlations with the HBG mRNA levels. Plasma levels of cytokines, such as erythropoietin, stem cell factor and transforming growth factor-beta were increased in patients, and these cytokines induced both HBG mRNA expression and CREB phosphorylation. These results demonstrate that the cAMP-dependent pathway, the activity of which is augmented by multiple cytokines, plays a role in regulating HBG expression in beta-thalassaemia.
Collapse
Affiliation(s)
- Lakiea Bailey
- Department of Medicine, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Costa FC, da Cunha AF, Fattori A, de Sousa Peres T, Costa GGL, Machado TF, de Albuquerque DM, Gambero S, Lanaro C, Saad STO, Costa FF. Gene expression profiles of erythroid precursors characterise several mechanisms of the action of hydroxycarbamide in sickle cell anaemia. Br J Haematol 2007; 136:333-42. [PMID: 17156400 DOI: 10.1111/j.1365-2141.2006.06424.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hydroxycarbamide (HC) (or hydroxyurea) has been reported to increase fetal haemoglobin levels and improve clinical symptoms in sickle cell anaemia (SCA) patients. However, the complete pathway by which HC acts remains unclear. To study the mechanisms involved in the action of HC, global gene expression profiles were obtained from the bone marrow cells of a SCA patient before and after HC treatment using serial analysis of gene expression. In the comparison of both profiles, 147 differentially expressed transcripts were identified. The functional classification of these transcripts revealed a group of gene categories associated with transcriptional and translational regulation, e.g. EGR-1, CENTB1, ARHGAP4 and RIN3, suggesting a possible role for these pathways in the improvement of clinical symptoms of SCA patients. The genes involved in these mechanisms may represent potential tools for the identification of new targets for SCA therapy.
Collapse
Affiliation(s)
- Flávia Chagas Costa
- The Haematology and Haemotherapy Centre, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Keefer JR, Schneidereith TA, Mays A, Purvis SH, Dover GJ, Smith KD. Role of cyclic nucleotides in fetal hemoglobin induction in cultured CD34+ cells. Exp Hematol 2006; 34:1151-61. [PMID: 16939808 DOI: 10.1016/j.exphem.2006.03.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 01/23/2006] [Accepted: 03/23/2006] [Indexed: 11/29/2022]
Abstract
OBJECTIVE In vivo, several drugs have been shown to increase fetal hemoglobin (HbF), including 5-azacytidine (AZA), sodium butyrate (SB), and hydroxyurea (HU). Studies in K562 cells suggest that cyclic guanosine monophosphate (cGMP) is required for HbF induction; however, the role of cyclic nucleotides in HbF induction in primary erythroid cultures has not been established. METHODS CD34-selected peripheral blood monocytes cultured in a semi-solid serum-free system that mimics in vivo F-cell production are utilized to explore the role of cyclic adenosine monophosphate (cAMP) and cGMP in HbF induction in response to HU, AZA, and SB. RESULTS In serum-free CD34 cultures, HU, SB, and AZA all markedly stimulate FNRBC production up to 30-fold, associated with induction of gamma-globin mRNA and total HbF protein. Guanylate cyclase inhibition results in only minimal blunting of HbF induction by each agent. In contrast, adenylate cyclase inhibition markedly reduces HU, SB, and AZA-mediated FNRBC induction and gamma-globin mRNA induction. The adenylate cyclase activator forskolin modestly induces FNRBC production and augments the action of standard induction agents. HU, AZA, and SB, however, fail to significantly stimulate adenylate cyclase themselves. CONCLUSIONS In human CD34(+) cultures, cAMP production is required for full induction of HbF by HU, SB, and AZA, while perturbation of cGMP production has only minimal effects. These findings are in marked contrast to data in K562 cells where cGMP production is critical for HbF induction while cAMP stimulation blunts HbF response, and suggest that these agents may share a common induction pathway.
Collapse
Affiliation(s)
- Jeffrey R Keefer
- Division of Pediatric Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Fathallah H, Atweh GF. DNA hypomethylation therapy for hemoglobin disorders: Molecular mechanisms and clinical applications. Blood Rev 2006; 20:227-34. [PMID: 16513230 DOI: 10.1016/j.blre.2006.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reactivation of fetal hemoglobin (HbF) expression is an important therapeutic option in patients with hemoglobin disorders. In sickle cell disease (SCD), an increase in HbF would interfere with the polymerization of sickle hemoglobin while in beta-thalassemia, an increase in gamma-globin chain synthesis would decrease non-alpha:alpha chain imbalance. Hydroxyurea, an inducer of HbF, is the only currently approved agent for the treatment of patients with moderate and/or severe SCD. However, about one third of patients with SCD do not respond to HU, and in beta-thalassemia, the clinical response is unimpressive. The last decade has seen a renewed interest in the use of inhibitors of DNA methylation in the treatment of patients with hemoglobin disorders. In this review, we discuss the role of DNA methylation in gamma-globin gene regulation, describe clinical trials with agents that hypomethylate DNA and speculate about the future role of DNA hypomethylation therapy in patients with SCD and beta-thalassemia.
Collapse
Affiliation(s)
- Hassana Fathallah
- Division of Hematology and Oncology, Box 1079, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA.
| | | |
Collapse
|
45
|
Hsiao CH, Li W, Lou TF, Baliga BS, Pace BS. Fetal hemoglobin induction by histone deacetylase inhibitors involves generation of reactive oxygen species. Exp Hematol 2006; 34:264-73. [PMID: 16543060 DOI: 10.1016/j.exphem.2005.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Revised: 11/28/2005] [Accepted: 12/08/2005] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Several compounds, including butyrate and trichostatin A, have been shown to activate gamma-gene expression via p38 mitogen-activated protein kinase (MAPK) signaling. In eukaryotic cells, reactive oxygen species (ROS) act as signaling molecules to mediate phosphorylation of tyrosine kinases such as p38 MAPK to regulate gene expression. Therefore, we determined the role of the reactive oxygen species hydrogen peroxide (H(2)O(2)) in drug-mediated fetal hemoglobin (HbF) induction. METHODS H(2)O(2) levels were measured using 2',7'-dichlorofluorescein-diacetate in K562 cells after drug treatments. To confirm a role for H(2)O(2) in HbF induction, studies were completed with the mitochondrial respiratory chain inhibitor myxothiazole, which prevents ROS generation. The ability of myxothiazole to block gamma-globin mRNA accumulation and HbF induction was measured in K562 cells and burst-forming unit-erythroid colonies respectively using quantitative real-time PCR and alkaline denaturation. RESULTS Butyrate and trichostastin A stimulated p38 MAPK phosphorylation via a H(2)O(2)-dependent mechanism. Pretreatment with myxothiazole to inhibit ROS formation or SB203580 to impede p38 MAPK signaling attenuated gamma-gene activation in K562 cells and HbF induction in erythroid progenitors. However, myxothiazole had no effect on the ability of hydroxyurea to induce HbF. CONCLUSION The findings presented herein support a ROS-p38 MAPK cell signaling mechanism for HbF induction by butyrate and trichostatin A.
Collapse
Affiliation(s)
- Cheng-Hui Hsiao
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX 75083, USA
| | | | | | | | | |
Collapse
|
46
|
Wolk M, Martin JE, Reinus C. Development of fetal haemoglobin-blood cells (F cells) within colorectal tumour tissues. J Clin Pathol 2006; 59:598-602. [PMID: 16469830 PMCID: PMC1860403 DOI: 10.1136/jcp.2005.029934] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AIM To evaluate the sources of fetal haemoglobin (HbF) as an indicator in cancer. An immunohistochemical study was carried out on some of the most common kinds of cancer. All of these cancers had serologically high levels of HbF as evaluated previously. METHODS Immunoaffinity-purified anti-HbF was immunohistochemically used to study F cell distribution in the following cancers: colorectal adenocarcinoma, urinary bladder transitional cell carcinoma, brain tumours, lung carcinoma, breast adenocarcinoma, leukaemia, Burkitt's lymphoma and endometrial carcinoma. RESULTS In colorectal adenocarcinoma, HbF-containing red blood cells (FRBC) were present within thin-walled vessels or were disposed in dense clusters within the tumour. Some of these cells were nucleated or binucleated HbF-erythroblasts or HbF-normoblasts (FNBS). In two cases, high levels of mitoses within HbF-erythroblasts were observed. In half of the cases with transitional cell carcinoma of the urinary bladder, regional intratumoral blood vessels were found to contain 5-50% FRBC. In the other tumours examined, F cells were not observed. FRBCs, however, were occasionally observed in the regional lymph nodes of some of these cancers. CONCLUSIONS The evaluation of HbF as a potential plasma marker is suggested by the high concentration of FRBCs in colorectal tumours. The apparent development of FRBCs in colorectal tumour tissues is an interesting observation, as these cells were previously thought to develop in medullary or lymphoid tissues. It is thus suggested that the colonic microenvironment may stimulate extramedullary fetal-type haematopoiesis.
Collapse
Affiliation(s)
- M Wolk
- Department of Morbid Anatomy and Histopathology, The Royal London Hospital, London, UK.
| | | | | |
Collapse
|
47
|
Iyamu EW, Cecil R, Parkin L, Woods G, Ohene-Frempong K, Asakura T. Modulation of erythrocyte arginase activity in sickle cell disease patients during hydroxyurea therapy. Br J Haematol 2006; 131:389-94. [PMID: 16225659 DOI: 10.1111/j.1365-2141.2005.05772.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An elevated erythrocyte arginase activity with a corresponding decrease in nitric oxide (NO) level has been implicated in the pathophysiology of sickle cell disease (SCD). Recent studies have shown that hydroxyurea (HU) increases the production of NO, which increases the soluble guanylate cyclase activity and fetal haemoglobin (HbF) synthesis. To study the effects of HU on the arginase and nitric oxide synthase (NOS) activities in SCD patients, we compared levels of arginase activity and NO metabolites in red blood cells and plasma, respectively, from 23 patients with SCD (HbSS) receiving HU therapy, with those of 12 SCD patients not receiving HU treatment. Patients on HU therapy showed significantly lower arginase activity than that of HbSS patients not on HU therapy (1.36+/-0.2 U/10(8) cells vs. 3.31+/-0.29 U/10(8) cells). NOS activity was higher in patients on HU therapy than in untreated patients (0.72+/-0.4 nmol/ml/min vs. 0.35+/-0.15 nmol/ml/min, P<0.05). Among the HU-treated patients, the decreased level of arginase activity correlated (r=0.71) with HbF level as well as the mean corpuscular haemoglobin content. These data suggest that one of the beneficial effects of HU in vivo may involve the regulation of arginase activity and a concomitant induction of NOS activity, which may lead to an increased production of NO. The outcome of this study may lead to the development of improved NO-based treatments for SCD.
Collapse
Affiliation(s)
- Efemwonkiekie W Iyamu
- Division of Hematology/Oncology, The Children's Mercy Hospital, Pediatric Research Center, Kansas City, MO 64108, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
The human globin genes are among the most extensively characterized in the human genome, yet the details of the molecular events regulating normal human hemoglobin switching and the potential reactivation of fetal hemoglobin in adult hematopoietic cells remain elusive. Recent discoveries demonstrate physical interactions between the beta locus control region and the downstream structural gamma- and beta-globin genes, and with transcription factors and chromatin remodeling complexes. These interactions all play roles in globin gene expression and globin switching at the human beta-globin locus. If the molecular events in hemoglobin switching were better understood and fetal hemoglobin could be more fully reactivated in adult cells, the insights obtained might lead to new approaches to the therapy of sickle cell disease and beta thalassemia by identifying specific new targets for molecular therapies.
Collapse
Affiliation(s)
- Arthur Bank
- Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
49
|
Tang DC, Zhu J, Liu W, Chin K, Sun J, Chen L, Hanover JA, Rodgers GP. The hydroxyurea-induced small GTP-binding protein SAR modulates gamma-globin gene expression in human erythroid cells. Blood 2005; 106:3256-63. [PMID: 15985540 PMCID: PMC1895330 DOI: 10.1182/blood-2003-10-3458] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hydroxyurea (HU), a drug effective in the treatment of sickle cell disease, is thought to indirectly promote fetal hemoglobin (Hb F) production by perturbing the maturation of erythroid precursors. The molecular mechanisms involved in HU-mediated regulation of gamma-globin expression are currently unclear. We identified an HU-induced small guanosine triphosphate (GTP)-binding protein, secretion-associated and RAS-related (SAR) protein, in adult erythroid cells using differential display. Stable SAR expression in K562 cells increased gamma-globin mRNA expression and resulted in macrocytosis. The cells appeared immature. SAR-mediated induction of gamma-globin also inhibited K562 cell growth by causing arrest in G1/S, apoptosis, and delay of maturation, cellular changes consistent with the previously known effects of HU on erythroid cells. SAR also enhanced both gamma- and beta-globin transcription in primary bone marrow CD34+ cells, with a greater effect on gamma-globin than on beta-globin. Although up-regulation of GATA-2 and p21 was observed both in SAR-expressing cells and HU-treated K562 cells, phosphatidylinositol 3 (PI3) kinase and phosphorylated ERK were inhibited specifically in SAR-expressing cells. These data reveal a novel role of SAR distinct from its previously known protein-trafficking function. We suggest that SAR may participate in both erythroid cell growth and gamma-globin production by regulating PI3 kinase/extracellular protein-related kinase (ERK) and GATA-2/p21-dependent signal transduction pathways.
Collapse
Affiliation(s)
- Delia C Tang
- Bldg 10, Rm 9N119, Molecular and Clinical Hematology Branch and Laboratory of Cell Biochemistry and Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Hydroxyurea is a relatively new treatment for sickle cell disease. A portion of hydroxyurea's beneficial effects may be mediated by nitric oxide, which has also drawn considerable interest as a sickle cell disease treatment. Patients taking hydroxyurea show a significant increase in iron nitrosyl hemoglobin and plasma nitrite and nitrate within 2 h of ingestion, providing evidence for the in vivo conversion of hydroxyurea to nitric oxide. Hydroxyurea reacts with hemoglobin to produce iron nitrosyl hemoglobin, nitrite, and nitrate, but these reactions do not occur fast enough to account for the observed increases in these species in patients taking hydroxyurea. This report reviews recent in vitro studies directed at better understanding the in vivo nitric oxide release from hydroxyurea in patients. Specifically, this report covers: (1) peroxidase-mediated formation of nitric oxide from hydroxyurea; (2) nitric oxide production after hydrolysis of hydroxyurea to hydroxylamine; and (3) the nitric oxide-producing structure-activity relationships of hydroxyurea. Results from these studies should provide a better understanding of the nitric oxide donor properties of hydroxyurea and guide the development of new hydroxyurea-derived nitric oxide donors as potential sickle cell disease therapies.
Collapse
Affiliation(s)
- S Bruce King
- Department of Chemistry, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|