1
|
Kotwal N, Pillai DK, Darbari DS, Sun K, Koumbourlis AC. Spirometric Changes After Initiation of Hydroxyurea in Children With Sickle Cell Anemia. J Pediatr Hematol Oncol 2022; 44:e923-e925. [PMID: 34966092 DOI: 10.1097/mph.0000000000002371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/25/2021] [Indexed: 11/25/2022]
Abstract
Individuals with sickle cell disease (SCD) develop a decline in lung function over time. Hydroxyurea (HU) is the most common disease-modifying therapy used in SCD. We hypothesized that children with SCD treated with HU will have a slower decline in pulmonary function. We performed a retrospective chart review of children with HbSS and HbS-beta zero thalassemia referred to pulmonology for respiratory symptoms. We compared the spirometry results at 2 time points between children on HU (HU group) and not on HU (control group). For the HU group, these endpoints were evaluated before and after being on HU. The mean time interval between 2 spirometry studies was not significantly different between the groups (2.6±1.5 y for HU group vs. 3.0±1.8 y for the control group; P =0.33). The mean age of patients in the HU group was 9.8±3.8 years (55% male) and 10.7±4.9 years (50% male) in the control group. The spirometry data was compared within and between the groups using t test. There was a significant increase in forced vital capacity in HU group during follow-up, while children in the control group showed a decline (7.2±17.1 vs. -3.4±18.2; P <0.01). Our study suggests that HU therapy may help preserve lung function over time in children with SCD.
Collapse
Affiliation(s)
- Nidhi Kotwal
- Department of Pediatrics, Division of Pulmonology and Allergy
- Division of Pulmonary and Sleep Medicine
| | | | - Deepika S Darbari
- Division of Hematology, Children's National Hospital, Washington, DC
| | - Kai Sun
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | | |
Collapse
|
2
|
Wamkpah N, Shrestha A, Salzman G, Simon S, Suman S, Poisner A, Molteni A. Renin-Angiotensin Blockade Reduces Readmission for Acute Chest Syndrome in Sickle Cell Disease. Cureus 2022; 14:e23567. [PMID: 35494947 PMCID: PMC9045847 DOI: 10.7759/cureus.23567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2022] [Indexed: 11/30/2022] Open
Abstract
Rationale Acute chest syndrome (ACS) is a life-threatening complication of sickle cell disease (SCD). Current treatment is supportive-supplemental oxygen, transfusions, and antibiotics. Prevention of ACS may reduce morbidity and mortality in patients with SCD. Acute chest syndrome appears similar to pulmonary fat embolism (PFE), a complication of severe skeletal trauma or orthopedic procedures from pulmonary micro-vessel blockage by bone marrow fat. Vascular obstruction and bone marrow necrosis occur in PFE and ACS. Pulmonary fat embolism rat models have shown that angiotensin-converting enzyme inhibitors (ACEI) and angiotensin II receptor blockers (ARB) mitigate damage in PFE. These medications could work similarly in ACS. We hypothesize that time to readmission after one hospitalization for ACS will be reduced in patients taking ACEI or ARB compared to patients who are not. Methods This is a retrospective cohort study. Inclusion criteria are adults (18 to 100 years) with sickle cell anaemia (HbSS), hemoglobin SC (HbSC) disease, sickle cell thalassemia (HbSβThal), hospitalized with ACS over 16 years (January 1, 2000, to March 31, 2016); patients who take and don’t take ACEI or ARB. Children (<18 years old), elderly adults (>100 years old), pregnant patients, and patients with sickle cell trait were excluded. Data was collected from the Health Facts database, which contains de-identified information from the electronic medical records of hospitals in which Cerner© has a data use agreement. Kaplan-Meier estimates explored a time-to-event model of ACS readmission. Multivariable analysis (age, gender, smoking history) was conducted using Cox proportional hazards regression. Results were reported around a 95% confidence interval. Results There were 6972 patients in total. Of which, 9.6% (n = 667) reported taking ACEI or ARB. Results for the covariates were: average age of 38 years old; 63% female (n = 4366/6969); 16% smokers (n = 1132). Readmission rates were higher for patients not taking ACEI/ARB than those who did: 0.44 (95% CI 0.43, 0.46) versus 0.28 (95% CI 0.24, 0.31) at one year, and 0.56 (95% CI 0.55, 0.58) versus 0.33 (95% CI 0.29, 0.37) at two years. Age had the strongest effect on readmission rates for patients taking ACEI/ARB (adjusted hazards ratio 0.78 [95% CI 0.68, 0.91]). Conclusion Patients with SCD who reported taking ACEI or ARB had lower readmission rates for ACS; age was the strongest covariate. Our results may have a significant impact on the prevention of ACS. Prospective studies comparing ACEI or ARB therapy versus placebo are needed to confirm this preventative effect.
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: 3.0] [Reference Citation Analysis] [Abstract] [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.
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
|
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
|
5
|
Shilo NR, Morris CR. Pathways to pulmonary hypertension in sickle cell disease: the search for prevention and early intervention. Expert Rev Hematol 2017; 10:875-890. [PMID: 28817980 DOI: 10.1080/17474086.2017.1364989] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Pulmonary hypertension (PH) develops in a significant number of patients with sickle cell disease (SCD), resulting in increased morbidity and mortality. This review focuses on PH pathophysiology, risk stratification, and new recommendations for screening and treatment for patients with SCD. Areas covered: An extensive PubMed literature search was performed. While the pathophysiology of PH in SCD is yet to be fully deciphered, it is known that the etiology is multifactorial; hemolysis, hypercoagulability, hypoxemia, ischemic-reperfusion injury, oxidative stress, and genetic susceptibility all contribute in varying degrees to endothelial dysfunction. Hemolysis, in particular, seems to play a key role by inciting an imbalance in the regulatory axis of nitric oxide and arginine metabolism. Systematic risk stratification starting in childhood based on clinical features and biomarkers that enable early detection is necessary. Multi-faceted, targeted interventions, before irreversible vasculopathy develops, will allow for improved patient outcomes and life expectancy. Expert commentary: Despite progress in our understanding of PH in SCD, clinically proven therapies remain elusive and additional controlled clinical trials are needed. Prevention of disease starts in childhood, a critical window for intervention. Given the complex and multifactorial nature of SCD, patients will ultimately benefit from combination therapies that simultaneously targets multiple mechanisms.
Collapse
Affiliation(s)
- Natalie R Shilo
- a Department of Pediatrics, Division of Pulmonary Medicine , University of Connecticut Heath Center , Farmington , CT , USA
| | - Claudia R Morris
- b Department of Pediatrics, Division of Pediatric Emergency Medicine, Emory-Children's Center for Cystic Fibrosis and Airways Disease Research , Emory University School of Medicine , Atlanta , GA , USA
| |
Collapse
|
6
|
da Guarda CC, Santiago RP, Pitanga TN, Santana SS, Zanette DL, Borges VM, Goncalves MS. Heme changes HIF-α, eNOS and nitrite production in HUVECs after simvastatin, HU, and ascorbic acid therapies. Microvasc Res 2016; 106:128-36. [PMID: 27089822 DOI: 10.1016/j.mvr.2016.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/09/2016] [Accepted: 04/07/2016] [Indexed: 12/31/2022]
Abstract
The sickle cell disease (SCD) is a hemolytic genetic anemia characterized by free heme and hemoglobin release into intravascular spaces, with endothelial activation. Heme is a proinflammatory molecule able to directly activate vascular endothelium, thus, endothelial dysfunction and vascular disease are major chronic events described in SCD. The aim of this study was to evaluate the production of endothelial nitric oxide synthase (eNOS), nitrite and hypoxia inducible factor alpha (HIF-α) in HUVECs (human umbilical vein endothelial cells) activated by heme in response to simvastatin, hydroxyurea (HU), and ascorbic acid therapies. eNOS and HIF-α production were evaluated by ELISA and nitrite was measured by the Griess technique. The production of HIF-α increased when the cells were stimulated by heme (p<0.01), while treatment with HU and simvastatin reduced the production (p<0.01), and treatment with ascorbic acid increased HIF-1a production by the cells (p<0.01). Heme increased eNOS production, (p<0.01) but showed a heterogeneous pattern, and the lowest concentrations of all the treatments reduced the enzyme production (p<0.01). The nitrite production by HUVECs was enhanced by stimulation with heme (p<0.001) and was reduced by treatment with HU (p<0.001), ascorbic acid (p<0.001) and simvastatin (p<0.01). In summary, our results suggest that the hemolytic vascular microenvironment in SCD requires different therapeutic approaches to promote clinical improvement, and that a combination of therapies may be a viable strategy for treating patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marilda S Goncalves
- Centro de Pesquisa Gonçalo Moniz/FIOCRUZ, Salvador, Brazil; Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil.
| |
Collapse
|
7
|
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: 13] [Impact Index Per Article: 1.6] [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
|
8
|
Goldstein S, Samuni A. Oxidation Mechanism of Hydroxamic Acids Forming HNO and NO. ADVANCES IN INORGANIC CHEMISTRY 2015. [DOI: 10.1016/bs.adioch.2014.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
9
|
Taira J, Tsuchida E, Katoh MC, Uehara M, Ogi T. Antioxidant capacity of betacyanins as radical scavengers for peroxyl radical and nitric oxide. Food Chem 2014; 166:531-536. [PMID: 25053090 DOI: 10.1016/j.foodchem.2014.05.102] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 05/12/2014] [Accepted: 05/17/2014] [Indexed: 11/30/2022]
Abstract
This study was designed to assess the antioxidant capacity of betacyanins as indole derived plant pigments, such as betanin, phyllocactin and betanidin. The antioxidant capacity of the betacyanins was evaluated as an index of radical scavenging ability using the peroxyl radical generating system in the presence of AAPH and NO generating system using NOR3 as an NO donor. The peroxyl radical scavenging capacity was dose-dependent in the low concentration range (25-100 nM). The mol-Trolox equivalent activity/mol compound (mol-TEA/mol-compound) as an index of the antioxidant capacity indicated the following order at 10.70 ± 0.01, 3.31 ± 0.14 and 2.83 ± 0.01 mol-TEA/mol-compound for betanidin, betanin and phyllocactin, respectively. In addition, betacyanins reduced the nitrite-level in the low concentration range of 2.5-20 μM. The IC₅₀ values (μM) of nitrogen radical scavenging activity were 24.48, 17.51 and 6.81 for betanin, phyllocactin and betanidin. ESR studies provided evidence that the compounds directly scavenged NO. These results indicated that betacyanins have a strong antioxidant capacity, particularly betanidin with a catechol group had higher activity than those of the glycoside of betacyanins. This study demonstrated that the betacyanins will be useful as natural pigments to provide defence against oxidative stress.
Collapse
Affiliation(s)
- Junsei Taira
- Department of Bioresource Technology, Okinawa National College of Technology, 905 Henoko, Nago City, Okinawa 905-2192, Japan.
| | - Eito Tsuchida
- Department of Bioresource Technology, Okinawa National College of Technology, 905 Henoko, Nago City, Okinawa 905-2192, Japan
| | - Megumi C Katoh
- Department of Bioresource Technology, Okinawa National College of Technology, 905 Henoko, Nago City, Okinawa 905-2192, Japan
| | - Masatsugu Uehara
- Department of Bioresource Technology, Okinawa National College of Technology, 905 Henoko, Nago City, Okinawa 905-2192, Japan
| | - Takayuki Ogi
- Okinawa Industrial Technology Center, 12-2 Suzaki, Uruma City, Okinawa 904-2234, Japan
| |
Collapse
|
10
|
Banan M. Hydroxyurea treatment in β-thalassemia patients: to respond or not to respond? Ann Hematol 2013; 92:289-99. [PMID: 23318979 DOI: 10.1007/s00277-012-1671-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 12/29/2012] [Indexed: 01/03/2023]
Abstract
Hydroxyurea (HU) is a drug that induces fetal hemoglobin production. As a result, HU is widely used to treat β-thalassemia (β-thal) patients. However, the response of these patients to HU varies. Some β-thal patients respond favorably to treatment while others do not respond at all. HU has a number of side-effects and therefore its targeted prescription is beneficial. Hence, identifying the genetic determinants which lead to the differential HU response is important. This review summarizes recent findings which have shed light on this topic. Special emphasis is given to the mechanisms and genetic loci which may govern these differences. These findings have helped identify several single nucleotide polymorphisms which associate with the response to HU in both β-thal and sickle cell disease patients.
Collapse
Affiliation(s)
- Mehdi Banan
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Evin, Daneshjoo Blvd., Koodakyar St, Tehran, Iran.
| |
Collapse
|
11
|
Samuni Y, Samuni U, Goldstein S. The use of cyclic nitroxide radicals as HNO scavengers. J Inorg Biochem 2012; 118:155-61. [PMID: 23122928 DOI: 10.1016/j.jinorgbio.2012.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/11/2012] [Accepted: 10/04/2012] [Indexed: 11/18/2022]
Abstract
Reduction of cyclic stable nitroxides (RNO) by HNO to the respective hydroxylamines (RNO-H) has been demonstrated using EPR spectrometry. HNO shows low reactivity toward piperidine, pyrrolidine and nitronyl nitroxides with rate constants below 1.4 × 10(5)M(-1)s(-1) at pH 7.0, despite the high driving force for these reactions. The rate constants can be predicted assuming that the reactions take place via a concerted proton-electron transfer pathway and significantly low self-exchange rate constants for HNO/NO and RNO-H/RNO. NO does not react with piperidine and pyrrolidine nitroxides, but does add to HNO forming the highly oxidizing and moderately reducing hyponitrite radicals. In this work, the radicals are produced by pulse radiolysis and the rate constants of their reactions with 2,2,6,6,-tetramethylpiperidine-1-oxyl (TEMPO), 4-hydroxy-2,2,6,6-tetramethyl piperidine-1-oxyl (TEMPOL) and 3-carbamoyl-PROXYL have been determined at pH 6.8 to be (2.4 ± 0.2)× 10(6), (9.8 ± 0.2)× 10(5), (5.9 ± 0.5)× 10(5)M(-1)s(-1), respectively. This low reactivity implies that NO competes efficiently with these nitroxides for the hyponitrite radical. The ability of TEMPOL and 2-(4-carboxyphenyl)-4,4,5,5,-tetramethyl-imidazoline-1-oxyl-3-oxide (C-PTIO) to oxidize HNO and their different reactivity toward NO are used to quantify HNO formed via acetohydroxamic acid oxidation. The extent of TEMPOL or C-PTIO reduction was similar to the yield of HNO formed upon oxidation by ()OH under anoxia, but not by the metmyoglobin and H(2)O(2) reaction system where both nitroxides catalytically facilitate H(2)O(2) depletion and nitrite accumulation. In this system the conversion of C-PTIO into 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl (C-PTI) is a minor reaction, which does not provide any mechanistic insight.
Collapse
Affiliation(s)
- Yuval Samuni
- Oral and Maxillofacial Surgery, The Barzilai Medical Center, Ashkelon, Israel
| | | | | |
Collapse
|
12
|
Samuni Y, Samuni U, Goldstein S. The mechanism underlying nitroxyl and nitric oxide formation from hydroxamic acids. Biochim Biophys Acta Gen Subj 2012; 1820:1560-6. [PMID: 22634736 DOI: 10.1016/j.bbagen.2012.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/19/2012] [Accepted: 05/17/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND The pharmacological effects of hydroxamic acids (RC(O)NHOH, HX) are partially attributed to their ability to serve as HNO and/or NO donors under oxidative stress. Given the development and use of HXs as therapeutic agents, elucidation of the oxidation mechanism is needed for more educated selection of HX-based drugs. METHODS Acetohydroxamic and glycine-hydroxamic acids were oxidized at pH 7.0 by a continuous flux of radiolytically generated (·)OH or by metmyoglobin and H(2)O(2) reactions system. Gas chromatography and spectroscopic methods were used to monitor the accumulation of N(2)O, N(2), nitrite and hydroxylamine. RESULTS Oxidation of HXs by (·)OH under anoxia yields N(2)O, but not nitrite, N(2) or hydroxylamine. Upon the addition of H(2)O(2) to solutions containing HX and metmyoglobin, which is instantaneously and continuously converted into compound II, nitrite and, to a lesser extent, N(2)O are accumulated under both anoxia and normoxia. CONCLUSIONS Oxidation of HXs under anoxia by a continuous flux of (·)OH, which solely oxidizes the hydroxamate moiety to RC(O)NHO(·), forms HNO. This observation implies that bimolecular decomposition of RC(O)NHO(·) competes efficiently with unimolecular decomposition processes such as internal disproportionation, hydrolysis or homolysis. Oxidation by metmyoglobin/H(2)O(2) involves relatively mild oxidants (compounds I and II). Compound I reacts with HX forming RC(O)NHO(·) and compound II, which oxidizes HX, RC(O)NHO(·), HNO and NO. The latter reaction is the main source of nitrite. GENERAL SIGNIFICANCE HXs under oxidative stress release HNO, but can be considered as NO-donors provided that HNO oxidation is more efficient than its reaction with other biological targets.
Collapse
Affiliation(s)
- Yuval Samuni
- Oral and Maxillofacial Surgery, The Brazilai Medical Center, Ashkelon, Israel
| | | | | |
Collapse
|
13
|
Vankayala SL, Hargis JC, Woodcock HL. Unlocking the binding and reaction mechanism of hydroxyurea substrates as biological nitric oxide donors. J Chem Inf Model 2012; 52:1288-97. [PMID: 22519847 DOI: 10.1021/ci300035c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hydroxyurea is the only FDA approved treatment of sickle cell disease. It is believed that the primary mechanism of action is associated with the pharmacological elevation of nitric oxide in the blood; however, the exact details of this are still unclear. In the current work, we investigate the atomic level details of this process using a combination of flexible-ligand/flexible-receptor virtual screening coupled with energetic analysis that decomposes interaction energies. Utilizing these methods, we were able to elucidate the previously unknown substrate binding modes of a series of hydroxyurea analogs to hemoglobin and the concomitant structural changes of the enzyme. We identify a backbone carbonyl that forms a hydrogen bond with bound substrates. Our results are consistent with kinetic and electron paramagnetic resonance (EPR) measurements of hydroxyurea-hemoglobin reactions, and a full mechanism is proposed that offers new insights into possibly improving substrate binding and/or reactivity.
Collapse
Affiliation(s)
- Sai Lakshmana Vankayala
- Department of Chemistry and Center for Molecular Diversity in Drug Design, Discovery, and Delivery, University of South Floridar, Tampa, Florida 33620, USA
| | | | | |
Collapse
|
14
|
Jones-Carson J, Laughlin JR, Stewart AL, Voskuil MI, Vázquez-Torres A. Nitric oxide-dependent killing of aerobic, anaerobic and persistent Burkholderia pseudomallei. Nitric Oxide 2012; 27:25-31. [PMID: 22521523 DOI: 10.1016/j.niox.2012.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 03/23/2012] [Accepted: 04/04/2012] [Indexed: 11/16/2022]
Abstract
Burkholderia pseudomallei infections are fastidious to treat with conventional antibiotic therapy, often involving a combination of drugs and long-term regimes. Bacterial genetic determinants contribute to the resistance of B. pseudomallei to many classes of antibiotics. In addition, anaerobiosis and hypoxia in abscesses typical of melioidosis select for persistent populations of B. pseudomallei refractory to a broad spectrum of antibacterials. We tested the susceptibility of B. pseudomallei to the drugs hydroxyurea, spermine NONOate and DETA NONOate that release nitric oxide (NO). Our investigations indicate that B. pseudomallei are killed by NO in a concentration and time-dependent fashion. The cytoxicity of this diatomic radical against B. pseudomallei depends on both the culture medium and growth phase of the bacteria. Rapidly growing, but not stationary phase, B. pseudomallei are readily killed upon exposure to the NO donor spermine NONOate. NO also has excellent antimicrobial activity against anaerobic B. pseudomallei. In addition, persistent bacteria highly resistant to most conventional antibiotics are remarkably susceptible to NO. Sublethal concentrations of NO inhibited the enzymatic activity of [4Fe-4S]-cofactored aconitase of aerobic and anaerobic B. pseudomallei. The strong anti-B. pseudomallei activity of NO described herein merits further studies on the application of NO-based antibiotics for the treatment of melioidosis.
Collapse
Affiliation(s)
- Jessica Jones-Carson
- Department of Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | | | | | | | | |
Collapse
|
15
|
Vrček IV, Šakić D, Vrček V, Zipse H, Biruš M. Computational study of radicals derived from hydroxyurea and its methylated analogues. Org Biomol Chem 2012; 10:1196-206. [DOI: 10.1039/c1ob06594g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
16
|
Hydroxyurea therapy mobilises arachidonic Acid from inner cell membrane aminophospholipids in patients with homozygous sickle cell disease. J Lipids 2011; 2011:718014. [PMID: 21941660 PMCID: PMC3173880 DOI: 10.1155/2011/718014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/15/2011] [Indexed: 01/29/2023] Open
Abstract
The cytotoxic compound hydroxyurea (HU) is effective therapy for sickle cell disease. However, its effect on unsaturated membrane lipids is unknown. Red cell fatty acids were investigated in HU-treated (n = 19) and HU-untreated (n = 17) sickle cell patients and controls (n = 20). The HU-treated compared with the HU-untreated patients had lower arachidonic (AA) acid level in ethanolamine, physphoglycerids (EPG) (22.9 ± 1.2 versus 24.0 ± 1.1%, P < 0.05) serine SPG (22.13 ± 2.2 versus 24.9 ± 2.3%, P < 0.01) phosphoglycerides. The treated patients and controls had comparable levels of docosahexaenoic (DHA) and total n-3 fatty acids in EPG and choline phosphoglycerides (CPG). In contrast, the untreated group had significantly (P < 0.05) lower DHA and total n-3 compared with the controls in EPG (2.7 ± 0.4 versus 3.2 ± 0.6% and 4.6 ± 0.5 versus 5.2 ± 0.7%) and CPG (0.7 ± 0.2 versus 1.0 ± 0.2%
and 1.2 ± 0.2 versus 1.4 ± 0.3). HU is known to activate cytosolic phospholipase A2 and cyclooxygenase 2, and from this study, it appears to induce mobilisation of AA from the inner cell membrane EPG and SPG. Hence, eicosanoids generated from the released AA may play a role in clinical improvements which occur in HU-treated patients.
Collapse
|
17
|
Samuni A, Goldstein S. One-Electron Oxidation of Acetohydroxamic Acid: The Intermediacy of Nitroxyl and Peroxynitrite. J Phys Chem A 2011; 115:3022-8. [DOI: 10.1021/jp201796q] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Amram Samuni
- Department of Molecular Biology, Medical School and #Chemistry Institute, The Accelerator Laboratory, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Sara Goldstein
- Department of Molecular Biology, Medical School and #Chemistry Institute, The Accelerator Laboratory, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
18
|
Kovacic P. Hydroxyurea (therapeutics and mechanism): Metabolism, carbamoyl nitroso, nitroxyl, radicals, cell signaling and clinical applications. Med Hypotheses 2011; 76:24-31. [DOI: 10.1016/j.mehy.2010.08.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 08/03/2010] [Accepted: 08/08/2010] [Indexed: 10/19/2022]
|
19
|
Samuni Y, Flores-Santana W, Krishna MC, Mitchell JB, Wink DA. The inhibitors of histone deacetylase suberoylanilide hydroxamate and trichostatin A release nitric oxide upon oxidation. Free Radic Biol Med 2009; 47:419-23. [PMID: 19447172 PMCID: PMC2730666 DOI: 10.1016/j.freeradbiomed.2009.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/07/2009] [Accepted: 05/09/2009] [Indexed: 11/16/2022]
Abstract
Suberoylanilide hydroxamic acid (SAHA, vorinostat, Zolinza) is the lead compound of a new class of histone deacetylase (HDAC) inhibitors used as anticancer drugs that have been shown to affect multiple proteins associated with gene expression, cell proliferation, and migration. Studies have also demonstrated the essential role of the hydroxamate moiety of SAHA in HDAC inhibition. The ability of SAHA and its structural analog trichostatin A (TSA) to generate NO upon oxidation was tested directly, by spin trapping of NO using electron paramagnetic resonance spectroscopy, and also indirectly, via the determination of nitrite using the Griess assay. H2O2/metmyoglobin was used to oxidize SAHA and TSA. These studies demonstrate, for the first time, the release of NO from SAHA and its structural analog TSA. We tested the protective effects of SAHA, TSA, and valproic acid (VPA) in mammalian Chinese hamster V79 cells exposed to a bolus of H2O2 for 1 h and monitored the clonogenic cell survival. Both SAHA and TSA afforded significant cytoprotection when co-incubated with H2O2, whereas VPA was ineffective. These studies provide evidence for the release of NO by hydroxamate-containing HDAC inhibitors and their antioxidant effects. Such roles may be an added advantage of this class of HDAC agents used for epigenetic therapies in cancer.
Collapse
Affiliation(s)
- Yuval Samuni
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
20
|
Lou TF, Singh M, Mackie A, Li W, Pace BS. Hydroxyurea generates nitric oxide in human erythroid cells: mechanisms for gamma-globin gene activation. Exp Biol Med (Maywood) 2009; 234:1374-82. [PMID: 19657070 DOI: 10.3181/0811-rm-339] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hydroxyurea (HU) induces fetal hemoglobin synthesis through activation of cyclic guanine monophosphate (cGMP) signaling. Studies in sickle cell patients demonstrated increased circulating nitric oxide (NO) levels after oral HU treatment. However, the direct measurement of NO in erythroid cells and its role in fetal hemoglobin induction have not been defined. Therefore, we quantified the level of nitrate and nitrite (NOx) generated by HU in human erythroid progenitors in the presence of three nitric oxide synthase inhibitors (NOS), including N(G)-monomethyl-L-arginine (L-NMMA). In addition, cGMP levels were measured in the presence or absence of the pathway inhibitor 1H-(1,2,4)ox-adiazolo(4,3-a)quinoxalin-1-one, which blocks soluble guanylyl cyclase formation. HU treatment increased NOx levels and gamma-globin transcription in K562 and primary erythroid cells, which was augmented when HU was combined with L-NMMA. Pretreatment with the cGMP pathway inhibitor reversed gamma-gene activation by HU. These data demonstrate the direct stimulation of cellular NO and cGMP signaling in erythroid progenitors by HU as a possible mechanism for gamma-globin gene activation.
Collapse
Affiliation(s)
- Tzu-Fang Lou
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, Texas 75080, USA
| | | | | | | | | |
Collapse
|
21
|
|
22
|
Fawibe AE. Managing acute chest syndrome of sickle cell disease in an African setting. Trans R Soc Trop Med Hyg 2008; 102:526-31. [PMID: 18455745 DOI: 10.1016/j.trstmh.2008.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Revised: 03/17/2008] [Accepted: 03/19/2008] [Indexed: 11/27/2022] Open
Abstract
Despite the fact that acute chest syndrome contributes immensely to morbidity and mortality in patients with sickle cell anaemia, its exact aetiopathogenesis is very complex and not yet well understood. Therefore, a high index of suspicion is needed in its diagnosis, and appropriate treatment should be commenced as soon as possible to prevent lethal complications of this condition, especially in Nigeria where appropriate diagnostic and therapeutic facilities may not be readily available. This is very important, as it may even develop on hospital admission. There is a need to further investigate preventive measures such as the use of hydroxyurea and the newly introduced Nicosan, especially in those people with recurrent disease, in order to reduce both short- and long-term complications of this syndrome among sickle cell patients in Nigeria.
Collapse
Affiliation(s)
- A E Fawibe
- Department of Internal Medicine, Federal Medical Center, Bida, Niger State, Nigeria.
| |
Collapse
|
23
|
Wood KC, Hsu LL, Gladwin MT. Sickle cell disease vasculopathy: a state of nitric oxide resistance. Free Radic Biol Med 2008; 44:1506-28. [PMID: 18261470 DOI: 10.1016/j.freeradbiomed.2008.01.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Revised: 11/21/2007] [Accepted: 01/11/2008] [Indexed: 12/31/2022]
Abstract
Sickle cell disease (SCD) is a hereditary hemoglobinopathy characterized by microvascular vaso-occlusion with erythrocytes containing polymerized sickle (S) hemoglobin, erythrocyte hemolysis, vasculopathy, and both acute and chronic multiorgan injury. It is associated with steady state increases in plasma cell-free hemoglobin and overproduction of reactive oxygen species (ROS). Hereditary and acquired hemolytic conditions release into plasma hemoglobin and other erythrocyte components that scavenge endothelium-derived NO and metabolize its precursor arginine, impairing NO homeostasis. Overproduction of ROS, such as superoxide, by enzymatic (xanthine oxidase, NADPH oxidase, uncoupled eNOS) and nonenzymatic pathways (Fenton chemistry), promotes intravascular oxidant stress that can likewise disrupt NO homeostasis. The synergistic bioinactivation of NO by dioxygenation and oxidation reactions with cell-free plasma hemoglobin and ROS, respectively, is discussed as a mechanism for NO resistance in SCD vasculopathy. Human physiological and transgenic animal studies provide experimental evidence of cardiovascular and pulmonary resistance to NO donors and reduced NO bioavailability that is associated with vasoconstriction, decreased blood flow, platelet activation, increased endothelin-1 expression, and end-organ injury. Emerging epidemiological data now suggest that chronic intravascular hemolysis is associated with certain clinical complications: pulmonary hypertension, cutaneous leg ulcerations, priapism, and possibly stroke. New therapeutic strategies to limit intravascular hemolysis and ROS generation and increase NO bioavailability are discussed.
Collapse
Affiliation(s)
- Katherine C Wood
- Vascular Medicine Branch, National Heart Lung Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
24
|
Nahavandi M, Tavakkoli F, Millis RM, Wyche MQ, Habib MJ, Tavakoli N. Effects of hydroxyurea and L-arginine on the production of nitric oxide metabolites in cultures of normal and sickle erythrocytes. ACTA ACUST UNITED AC 2007; 11:291-4. [PMID: 17178670 DOI: 10.1080/10245330600921998] [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: 10/23/2022]
Abstract
Previous in vitro studies suggest that erythrocytes may be a source of nitric oxide (NO) produced by nitric oxide synthase (NOS) or by oxyhemoglobin-mediated oxidation of hydroxyurea (HU). This study was performed to determine the roles of HU and NOS in the production of NO by normal and sickle erythrocytes. Red blood cells (RBCs) from normal adult hemoglobin (HbAA) and homozygous sickle cell subjects (HbSS) were incubated with PBS containing 0.2 mM hydrogen peroxide (control) for 2 h at 37 degrees C in the presence and absence of l-arginine, the substrate for NOS, and with l-arginine plus HU in the presence and absence of l-NMMA, a specific inhibitor of NOS. The nitrate and nitrite metabolites of NO, expressed as [NOx], were measured. [NOx] in the HbAA and HbSS RBC cultures was not significantly different in the presence and absence of 1.0 mM l-arginine (p>0.1). [NOx] in the HbAA and HbSS cultures treated with a clinically relevant dose of HU (1.0 mM) plus 1.0 mM l-arginine was significantly greater than that in controls incubated with PBS and with l-arginine p < 0.01. However, [NOx] in the HbAA and HbSS cultures treated with 50 microg/ml l-NMMA was not significantly different than that in the cultures treated with HU plus l-arginine in the absence of l-NMMA. These findings suggest that NOx production by erythrocytes may be increased by treatment with HU and may not be decreased by inhibiting NOS. Therefore, we conclude that a therapeutic dose of HU may increase the plasma concentration of NO by a mechanism that does not require erythrocytes NOS activity.
Collapse
Affiliation(s)
- Masoud Nahavandi
- Departments of Anesthesiology, Howard University College of Medicine, Washington, DC 20059, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Steinberg MH. Pathophysiologically based drug treatment of sickle cell disease. Trends Pharmacol Sci 2006; 27:204-10. [PMID: 16530854 DOI: 10.1016/j.tips.2006.02.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Revised: 12/01/2005] [Accepted: 02/21/2006] [Indexed: 01/18/2023]
Abstract
Sickle cell disease is a systemic disorder that is caused by a mutation (Glu6Val) in the gene that encodes beta globin. The sickle hemoglobin molecule (HbS) is a tetramer of two alpha-globin chains and two sickle beta-globin chains, and has the tendency to polymerize when deoxygenated. HbS facilitates abnormal interactions between the sickle erythrocyte and leukocytes and endothelial cells, which trigger a complex pathobiology. This multifaceted pathophysiology provides the opportunity to interrupt the disease at multiple sites, including polymerization of HbS, erythrocyte density and cell-cell interactions. For example, it is possible to induce higher concentrations of fetal hemoglobin, which disrupts the pathology-initiating step of HbS polymerization. Furthermore, it is possible to improve the hydration of sickle erythrocytes and it might be feasible to counteract the endothelial, inflammatory and oxidative abnormalities of sickle cell disease. A therapeutic approach that targets several sites of pathobiology might be most promising.
Collapse
Affiliation(s)
- Martin H Steinberg
- Center of Excellence in Sickle Cell Disease, E248, Boston Medical Center, 88 E. Newton Street, Boston, MA 02118, USA.
| |
Collapse
|
26
|
Burkitt MJ, Raafat A. Nitric oxide generation from hydroxyurea: significance and implications for leukemogenesis in the management of myeloproliferative disorders. Blood 2006; 107:2219-22. [PMID: 16282342 DOI: 10.1182/blood-2005-08-3429] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe use of myelosuppressive agents to reduce the risk of thrombosis in patients with polycythemia vera (PV) and essential thrombocythemia (ET) has been associated with an increased risk of transformation to acute myeloid leukemia (AML). Whereas chlorambucil, busulfan, and radiophosphorus (32P) have been demonstrated to increase the risk of transformation, the leukemogenic potential of hydroxyurea (HU) continues to be a matter of debate. Clinical studies have suggested that HU may cause a small increase in the risk of AML, but it has proven difficult to establish whether AML is actually caused by HU or arises during the natural progression of PV and ET. Reports that HU undergoes metabolic activation to species that induce mutation appear to support the notion that it is leukemogenic. Here, we suggest that the ability of HU to induce mutation in cell culture studies results from the generation of nitrogen dioxide via the autoxidation of nitric oxide, a product of HU metabolism. However, we argue that autoxidation would not occur in vivo, leading to the conclusion that generation of the mutagen nitrogen dioxide is peculiar to cell culture systems and has little relevance to the use of HU in the management of PV and ET.
Collapse
Affiliation(s)
- Mark J Burkitt
- Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood, Middlesex, HA6 2JR United Kingdom.
| | | |
Collapse
|
27
|
Abstract
Priapism, although uncommon in the general population, is one of the many serious complications associated with sickle cell disease (SCD). Few studies have described the clinical and hematologic characteristics of individuals with priapism and SCD. Using data from the Cooperative Study for Sickle Cell Disease, we assembled 273 case subjects with priapism and 979 control subjects. Case subjects, compared with control subjects, had significantly lower levels of hemoglobin; higher levels of lactate dehydrogenase, bilirubin, and aspartate aminotransferase; and higher reticulocyte, white blood cell, and platelet counts. These findings suggest an association of priapism with increased hemolysis. Hemolysis decreases the availability of circulating nitric oxide, which plays an important role in erectile function.
Collapse
Affiliation(s)
- Vikki G Nolan
- Department of Medicine, Boston University School of Medicine, MA, USA
| | | | | | | |
Collapse
|
28
|
Nigović B, Kujundzić N, Sanković K. Electron transfer in N-hydroxyurea complexes with iron(III). Eur J Med Chem 2005; 40:51-5. [PMID: 15642408 DOI: 10.1016/j.ejmech.2004.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Revised: 07/22/2004] [Accepted: 09/08/2004] [Indexed: 11/24/2022]
Abstract
Redox behaviour of the iron(III) complex with the antitumour drug hydroxyurea was studied by cyclic voltammetry. The complex underwent a one-electron reduction, followed by an irreversible chemical reaction (EC mechanism) in which a ligand was released. In addition, it was found that the hydroxyurea gave up an electron to iron(III) in solution. Differential-pulse voltammetry revealed an increase in the concentration of the generated iron(II) species. Electron paramagnetic resonance (EPR) spectroscopy studies of the oxidative degradation of hydroxyurea confirmed formation of the radical species H2N-CO-NHO*. Electrochemical data for iron(III) complexes of hydroxyurea and its structural analogue 3-ethylhydroxyurea, which also exhibits antitumour activity, show the same mechanism involved in the electron transfer. The observed redox properties indicate that hydroxyurea may interfere with electron transfer processes in biological systems after binding to iron-containing ribonucleotide reductase.
Collapse
Affiliation(s)
- Biljana Nigović
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia.
| | | | | |
Collapse
|
29
|
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
|
30
|
Haynes J, Baliga BS, Obiako B, Ofori-Acquah S, Pace B. Zileuton induces hemoglobin F synthesis in erythroid progenitors: role of the L-arginine-nitric oxide signaling pathway. Blood 2004; 103:3945-50. [PMID: 14764535 DOI: 10.1182/blood-2003-08-2969] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Induction of fetal hemoglobin (Hb F) is an important therapeutic tool in ameliorating complications of sickle cell disease. Nitric oxide has been implicated in the mechanism of Hb F synthesis induced by hydroxyurea (HU). This study examined whether zileuton (ZL), a structural analog of hydroxyurea, possessed Hb F-inducing properties and the potential role nitric oxide plays. ZL caused a dose-dependent increase in gamma-globin expression in K562 cells. This effect was confirmed by a dose-dependent increase in Hb F synthesis in erythroid progenitors from individuals with sickle cell anemia and normal hemoglobin genotypes. l-arginine had no effect on Hb F production; however, it dose-dependently inhibited ZL's ability to induce Hb F. The nitric oxide synthase inhibitor N(G)-monomethyl-l-arginine (l-NMMA) inhibited l-arginine's effect and restored ZL-mediated increase in Hb F synthesis. In addition, 8-PCPT-cGMP (8-(4-chlorophenylthio)guanosine 3',5'-cyclic monophosphate) inhibited ZL-mediated induction of Hb F synthesis. When comparing l-NMMA effects alone on ZL and HU, a partial reversal of increased Hb F synthesis was seen only with HU. Neither l-arginine alone nor l-arginine in combination with l-NMMA effected hydroxyurea-mediated induction of Hb F synthesis. This study demonstrates that ZL induces Hb F through a mechanism that involves l-arginine/nitric oxide/cGMP in a manner distinctly different from HU.
Collapse
Affiliation(s)
- Johnson Haynes
- Department of Medicine, University of South Alabama Medical Center, Mobile, AL 36617, USA.
| | | | | | | | | |
Collapse
|
31
|
Huang J, Zou Z, Kim-Shapiro DB, Ballas SK, King SB. Hydroxyurea analogues as kinetic and mechanistic probes of the nitric oxide producing reactions of hydroxyurea and oxyhemoglobin. J Med Chem 2003; 46:3748-53. [PMID: 12904079 DOI: 10.1021/jm0301538] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Derivatives of N-hydroxyurea that contain an N-hydroxy group react with oxyhemoglobin to form methemoglobin and variable amounts of nitrite/nitrate. Compounds with an unsubstituted -NHOH group produce the most nitrite/nitrate, which provides evidence for nitric oxide formation. The rate of reaction of these N-hydroxyurea derivatives with oxyhemoglobin correlates well with that compound's oxidation potential. Aromatic N-hydroxyureas react 25-80-fold faster with oxyhemoglobin than with N-hydroxyurea, suggesting other N-hydroxyurea analogues may be superior nitric oxide donors. Electron paramagnetic resonance spectroscopy shows that the formation of a low-spin methemoglobin-hydroxyurea complex is critical for iron nitrosyl hemoglobin formation. These results show that iron nitrosyl hemoglobin formation from the reaction of hydroxyureas and hemoglobin requires an unsubstituted -NHOH group and that the nitrogen atom of the non-N-hydroxy group must contain at least a single hydrogen atom. These results should guide the development of new hydroxyurea-based nitric oxide donors and sickle cell disease therapies.
Collapse
Affiliation(s)
- Jinming Huang
- Departments of Chemistry and Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | | | | | | | | |
Collapse
|
32
|
Mak V, Davies SC. The pulmonary physician in critical care * Illustrative case 6: Acute chest syndrome of sickle cell anaemia. Thorax 2003; 58:726-8. [PMID: 12885995 PMCID: PMC1746766 DOI: 10.1136/thorax.58.8.726] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- V Mak
- Department of Respiratory and Critical Care Medicine, Central Middlesex Hospital, London NW10 7NS, UK.
| | | |
Collapse
|
33
|
Lockamy VL, Huang J, Shields H, Ballas SK, King SB, Kim-Shapiro DB. Urease enhances the formation of iron nitrosyl hemoglobin in the presence of hydroxyurea. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1622:109-16. [PMID: 12880948 DOI: 10.1016/s0304-4165(03)00132-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although it has been shown that hydroxyurea (HU) therapy produces measurable amounts of nitric oxide (NO) metabolites, including iron nitrosyl hemoglobin (HbNO) in patients with sickle cell disease, the in vivo mechanism for formation of these is not known. Much in vitro data and some in vivo data indicates that HU is the NO donor, but other studies suggest a role for nitric oxide synthase (NOS). In this study, we confirm that the NO-forming reactions of HU with hemoglobin (Hb) or other blood constituents is too slow to account for NO production measured in vivo. We hypothesize that, in vivo, HU is partially metabolized to hydroxylamine (HA), which quickly reacts with Hb to form methemoglobin (metHb) and HbNO. We show that addition of urease, which converts HU to HA, to a mixture of blood and HU, greatly enhances HbNO formation.
Collapse
Affiliation(s)
- Virginia L Lockamy
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109-7507, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ho JA, Pickens CV, Gamcsik MP, Colvin OM, Ware RE, Gamscik MP. In vitro induction of fetal hemoglobin in human erythroid progenitor cells. Exp Hematol 2003; 31:586-91. [PMID: 12842703 DOI: 10.1016/s0301-472x(03)00086-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Clinical heterogeneity among patients with sickle cell anemia (SCA) is influenced by the amount of fetal hemoglobin (HbF) within circulating erythrocytes. Current pharmacotherapy focuses on increasing HbF in order to reduce hemolysis and help prevent acute vaso-occlusive events. Hydroxyurea, a known S-phase-specific cytotoxic ribonucleotide reductase (RR) inhibitor, is an effective agent for HbF induction in patients with SCA, but the mechanisms by which hydroxyurea induces HbF in vivo have not been elucidated. MATERIALS AND METHODS We adapted an in vitro assay for HbF induction, growing burst-forming unit erythroid (BFU-E) colonies in methylcellulose from peripheral blood of children with SCA and extracting the hemoglobin for high-performance liquid chromatography analysis of HbF. Hydroxyurea and other known RR inhibitors, along with cytotoxic agents that are not RR inhibitors, were tested for the ability to induce HbF using this in vitro assay. RESULTS Hydroxyurea decreased the number of BFU-E colonies that grew in culture and significantly increased HbF from 13.6%+/-6.2% to 25.4%+/-8.0% at 50 microM HU (p=0.012). Three other known RR inhibitors also significantly induced HbF: 4-methyl-5-amino-1-formylisoquinoline thiosemicarbazone (p=0.025), guanazole (p=0.008), and gemcitabine (p=0.028). Cytarabine and alkylating agents BCNU and 4-hydroperoxycyclophosphamide, which are cytotoxic agents but not RR inhibitors, reduced BFU-E colony number but did not significantly induce HbF. CONCLUSION Hydroxyurea and other RR inhibitors significantly induce HbF in vitro in human erythroid progenitor cells. Inhibition of RR may be a critical mechanism by which hydroxyurea increases HbF in vivo in patients with SCA.
Collapse
Affiliation(s)
- Janie A Ho
- Division of Pediatric Hematology/Oncology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Sickle cell disease (SCD) is one of the most common genetic diseases with some 250,000 new births each year. Most patients suffer intermittent pain crises and life-threatening events while life expectancy is considerably reduced. Until the last decade management was purely preventative or supportive aimed at symptom control. Apart from stem cell transplant, there is no cure but the oral chemotherapeutic drug hydroxyurea (HU) has now established a role in ameliorating the disease and improving life expectancy for most patients. There are side effects and risks of HU treatment in SCD but for moderate and severely affected patients, the benefits can be significant.
Collapse
Affiliation(s)
- Sally C Davies
- Imperial College Faculty of Medicine at Central Middlesex Hospital, Acton Lane, London NW10 7NS, UK.
| | | |
Collapse
|
36
|
Reiter CD, Gladwin MT. An emerging role for nitric oxide in sickle cell disease vascular homeostasis and therapy. Curr Opin Hematol 2003; 10:99-107. [PMID: 12579034 DOI: 10.1097/00062752-200303000-00001] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nitric oxide participates in the compensatory response to chronic vascular injury in patients with sickle cell disease. The authors have found reductions of basal and stimulated nitric oxide production and responses to exogenous nitric oxide in male patients with sickle cell disease. Gender differences in nitric oxide bioavailability are probably caused in part by the protective effects of ovarian estrogen on nitric oxide synthase expression and activity in women. Further, in men, and likely all patients during vaso-occlusive crisis and the acute chest syndrome, nitric oxide is destroyed by increased circulating plasma hemoglobin and superoxide. The combined effects of inhaled nitric oxide gas of improving pulmonary ventilation to perfusion matching and hemodynamics, reducing alveolar and systemic inflammation, and inhibiting circulating plasma hemoglobin (and thus restoring peripheral nitric oxide bioavailability) may modulate the course of the disease, including the frequency and severity of vaso-occlusive crises and acute chest syndrome episodes. Possible effects of chronic nitric oxide-based therapies on erythrocyte density, pulmonary artery pressures, and fetal hemoglobin induction deserve study.
Collapse
Affiliation(s)
- Christopher D Reiter
- Critical Care Medicine Department, Warren G Magnuson Clinical Center, National Institutes of Health, Bethesda, Maryland 20892-1662, USA
| | | |
Collapse
|
37
|
King SB. A role for nitric oxide in hydroxyurea-mediated fetal hemoglobin induction. J Clin Invest 2003; 111:171-2. [PMID: 12531869 PMCID: PMC151883 DOI: 10.1172/jci17597] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- S Bruce King
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina 27109, USA.
| |
Collapse
|
38
|
Cokic VP, Smith RD, Beleslin-Cokic BB, Njoroge JM, Miller JL, Gladwin MT, Schechter AN. Hydroxyurea induces fetal hemoglobin by the nitric oxide–dependent activation of soluble guanylyl cyclase. J Clin Invest 2003. [DOI: 10.1172/jci200316672] [Citation(s) in RCA: 233] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
39
|
King SB. A role for nitric oxide in hydroxyurea-mediated fetal hemoglobin induction. J Clin Invest 2003. [DOI: 10.1172/jci200317597] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
40
|
Cokic VP, Smith RD, Beleslin-Cokic BB, Njoroge JM, Miller JL, Gladwin MT, Schechter AN. Hydroxyurea induces fetal hemoglobin by the nitric oxide-dependent activation of soluble guanylyl cyclase. J Clin Invest 2003; 111:231-9. [PMID: 12531879 PMCID: PMC151872 DOI: 10.1172/jci16672] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hydroxyurea treatment of patients with sickle-cell disease increases fetal hemoglobin (HbF), which reduces hemoglobin S polymerization and clinical complications. Despite its use in the treatment of myeloproliferative diseases for over 30 years, its mechanism of action remains uncertain. Recent studies have demonstrated that hydroxyurea generates the nitric oxide (NO) radical in vivo, and we therefore hypothesized that NO-donor properties might determine the hemoglobin phenotype. We treated both K562 erythroleukemic cells and human erythroid progenitor cells with S-nitrosocysteine (CysNO), an NO donor, and found similar dose- and time-dependent induction of gamma-globin mRNA and HbF protein as we observed with hydroxyurea. Both hydroxyurea and CysNO increased cGMP levels, and the guanylyl cyclase inhibitors ODQ, NS 2028, and LY 83,538 abolished both the hydroxyurea- and CysNO-induced gamma-globin expression. These data provide strong evidence for an NO-derived mechanism for HbF induction by hydroxyurea and suggest possibilities for therapies based on NO-releasing or -potentiating agents.
Collapse
Affiliation(s)
- Vladan P Cokic
- Laboratory of Chemical Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Huang J, Sommers EM, Kim-Shapiro DB, King SB. Horseradish peroxidase catalyzed nitric oxide formation from hydroxyurea. J Am Chem Soc 2002; 124:3473-80. [PMID: 11916434 DOI: 10.1021/ja012271v] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hydroxyurea represents an approved treatment for sickle cell anemia and a number of cancers. Chemiluminescence and electron paramagnetic resonance spectroscopic studies show horseradish peroxidase catalyzes the formation of nitric oxide from hydroxyurea in the presence of hydrogen peroxide. Gas chromatographic headspace analysis and infrared spectroscopy also reveal the production of nitrous oxide in this reaction, which provides evidence for nitroxyl, the one-electron reduced form of nitric oxide. These reactions also generate carbon dioxide, ammonia, nitrite, and nitrate. None of these products form within 1 h in the absence of hydrogen peroxide or horseradish peroxidase. Electron paramagnetic resonance spectroscopy and trapping studies show the intermediacy of a nitroxide radical and a C-nitroso species during this reaction. Absorption spectroscopy indicates that both compounds I and II of horseradish peroxidase act as one-electron oxidants of hydroxyurea. Nitroxyl, generated from Angeli's salt, reacts with ferric horseradish peroxidase to produce a ferrous horseradish peroxidase-nitric oxide complex. Electron paramagnetic resonance experiments with a nitric oxide specific trap reveal that horseradish peroxidase is capable of oxidizing nitroxyl to nitric oxide. A mechanistic model that includes the observed nitroxide radical and C-nitroso compound intermediates has been forwarded to explain the observed product distribution. These studies suggest that direct nitric oxide producing reactions of hydroxyurea and peroxidases may contribute to the overall pharmacological properties of this drug.
Collapse
Affiliation(s)
- Jinming Huang
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina 27109, USA
| | | | | | | |
Collapse
|
42
|
Wang PG, Xian M, Tang X, Wu X, Wen Z, Cai T, Janczuk AJ. Nitric oxide donors: chemical activities and biological applications. Chem Rev 2002; 102:1091-134. [PMID: 11942788 DOI: 10.1021/cr000040l] [Citation(s) in RCA: 972] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Peng George Wang
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Taira J, Miyagi C, Aniya Y. Dimerumic acid as an antioxidant from the mold, Monascus anka: the inhibition mechanisms against lipid peroxidation and hemeprotein-mediated oxidation. Biochem Pharmacol 2002; 63:1019-26. [PMID: 11911855 DOI: 10.1016/s0006-2952(01)00923-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This study aimed to investigate the antioxidant mechanism of dimerumic acid isolated as the active component with a radical scavenging action from the mold Monascus anka, traditionally used for the fermentation of foods. Dimerumic acid inhibited NADPH- and iron(II)-dependent lipid peroxidation (LPO) of rat liver microsomes at 20 and 200 microM, respectively. When ferrylmyoglobin was incubated with dimerumic acid, the myoglobin was scavenged and an electron spin resonance (ESR) signal with nine peaks was observed. The spin adduct was identified as a nitroxide radical by analysis of hyperfine structure. Similar ESR signal was also detected by incubation of dimerumic acid with peroxyl radicals. Thus, it was clarified that the antioxidant action of dimerumic acid is due to one electron donation of the hydroxamic acid group in the dimerumic acid molecule toward oxidants resulting in formation of nitroxide radical.
Collapse
Affiliation(s)
- Junsei Taira
- Okinawa Prefectural Institute of Health and Environment, Okinawa, Japan
| | | | | |
Collapse
|
44
|
Huang J, Hadimani SB, Rupon JW, Ballas SK, Kim-Shapiro DB, King SB. Iron nitrosyl hemoglobin formation from the reactions of hemoglobin and hydroxyurea. Biochemistry 2002; 41:2466-74. [PMID: 11841242 DOI: 10.1021/bi011470o] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hydroxyurea represents an approved treatment for sickle cell anemia and acts as a nitric oxide donor under oxidative conditions in vitro. Electron paramagnetic resonance spectroscopy shows that hydroxyurea reacts with oxy-, deoxy-, and methemoglobin to produce 2-6% of iron nitrosyl hemoglobin. No S-nitrosohemoglobin forms during these reactions. Cyanide and carbon monoxide trapping studies reveal that hydroxyurea oxidizes deoxyhemoglobin to methemoglobin and reduces methemoglobin to deoxyhemoglobin. Similar experiments reveal that iron nitrosyl hemoglobin formation specifically occurs during the reaction of hydroxyurea and methemoglobin. Experiments with hydroxyurea analogues indicate that nitric oxide transfer requires an unsubstituted acylhydroxylamine group and that the reactions of hydroxyurea and deoxy- and methemoglobin likely proceed by inner-sphere mechanisms. The formation of nitrate during the reaction of hydroxyurea and oxyhemoglobin and the lack of nitrous oxide production in these reactions suggest the intermediacy of nitric oxide as opposed to its redox form nitroxyl. A mechanistic model that includes a redox cycle between deoxyhemoglobin and methemoglobin has been forwarded to explain these results that define the reactivity of hydroxyurea and hemoglobin. These direct nitric oxide producing reactions of hydroxyurea and hemoglobin may contribute to the overall pathophysiological properties of this drug.
Collapse
Affiliation(s)
- Jinming Huang
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina 27109, USA
| | | | | | | | | | | |
Collapse
|
45
|
Gladwin MT, Shelhamer JH, Ognibene FP, Pease-Fye ME, Nichols JS, Link B, Patel DB, Jankowski MA, Pannell LK, Schechter AN, Rodgers GP. Nitric oxide donor properties of hydroxyurea in patients with sickle cell disease. Br J Haematol 2002; 116:436-44. [PMID: 11841449 DOI: 10.1046/j.1365-2141.2002.03274.x] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hydroxyurea therapy reduces the rates of vaso-occlusive crisis in patients with sickle cell anaemia and recent data suggest that hydroxyurea treatment can generate nitric oxide (NO). Nitric oxide has been proposed as a novel therapy for sickle cell disease via a number of pathways. We therefore sought to determine whether hydroxyurea has NO donor properties in patients with sickle cell anaemia and explore potential mechanisms by which NO production could be therapeutic. Venous blood was collected from 19 fasting sickle cell anaemia patients, on chronic hydroxyurea therapy, at baseline and 2 and 4 h after a single morning dose of hydroxyurea, as well as 10 patients not taking hydroxyurea. The plasma and red cell NO reaction products nitrate, nitrite and nitrosylated- haemoglobin were measured using ozone-based chemiluminescent assays (using vanadium, KI and I3- reductants respectively). Consistent with NO release from hydroxyurea, baseline levels of total nitrosylated haemoglobin increased from 300 nmol/l to 500 nmol/l (P = 0.01). Plasma nitrate and nitrite levels also significantly increased with peak levels observed at 2 h. Glutathionyl-haemoglobin levels were unchanged, while plasma secretory vascular cellular adhesion molecule-1 levels were reduced in patients taking hydroxyurea (419 +/- 40 ng/ml) compared with control patients with sickle cell anaemia (653 +/- 55 ng/ml; P = 0.003), and were inversely correlated with fetal haemoglobin levels (r = -0.72; P = 0.002). These results demonstrate that hydroxyurea therapy is associated with the intravascular and intraerythrocytic generation of NO. The role of NO in the induction of fetal haemoglobin and possible synergy between NO donor therapy and classic cytostatic and differentiating medications should be explored.
Collapse
Affiliation(s)
- Mark T Gladwin
- Critical Care Medicine Department of the Warren G. Magnuson Clinical Center, Bethesda, MD 20892-1662, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Recent clinical and experimental data suggest that nitric oxide (NO) may play a role in the pathogenesis and therapy of sickle cell disease. NO, a soluble gas continuously synthesized in endothelial cells by the NO synthase (NOS) enzyme systems, regulates basal vascular tone and endothelial function, and maintains blood oxygenation via hypoxic pulmonary vasoconstriction and reduced shunt physiology. These vital homeostatic processes may be impaired in sickle cell disease and contribute to its pathogenesis. Therapeutic NO inhalation exerts significant direct effects on the pulmonary vasculature to reduce pulmonary pressures and increase oxygenation that may prove beneficial in acute chest syndrome and secondary pulmonary hypertension. Delivery of NO bound to hemoglobin or in plasma may improve blood flow and hemoglobin saturation, and thus reduce ischemia-reperfusion injury. Other NO-related effects on adhesion molecule expression and fetal hemoglobin induction are of interest. While direct evidence for a clinical benefit of NO therapy in sickle cell disease has not been reported, studies are underway to determine if inhaled NO will reduce the substantial morbidity and mortality suffered by these patients.
Collapse
Affiliation(s)
- M T Gladwin
- Critical Care Medicine Department, Warren G. Magnuson Clinical Center, NIH, Bethesda, MD 20892-1662, USA
| | | |
Collapse
|
47
|
Nahavandi M, Wyche MQ, Perlin E, Tavakkoli F, Castro O. Nitric Oxide Metabolites in Sickle Cell Anemia Patients after Oral Administration of Hydroxyurea; Hemoglobinopathy. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2001; 5:335-339. [PMID: 11399633 DOI: 10.1080/10245332.2000.11746528] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The mechanism of action of hydroxyurea (HU) in decreasing the frequency of pain crisis in sickle cell disease (SCD) has not been fully elucidated. In vitro and in vivo studies suggest that nitric oxide (NO), a potent vasodilator, may partly be responsible for the beneficial effect of HU. This study was designed to determine the effect of oral administration of HU on plasma levels of NO metabolites (NO(x) ) in sickle cell patients (SCP). The results indicate that during steady-state plasma levels of NO(x) were significantly higher in HU-treated patients compared to non HU-treated patients or normal controls (p <.05). In five inpatients in mild pain plasma levels of NO(x) increased significantly after 2 h of HU administration (p <.05); however, in three inpatients in persistent pain with significantly lower baseline NO(x) there was a minimal NO(x) response to HU at 2 h (p <.01). These observations indicate that HU administration is associated with the production of NO in some SCP, but that further study of the pharmacodynamics of this effect is necessary.
Collapse
Affiliation(s)
- Masoud Nahavandi
- Departments of Anesthesiology, Internal Medicine, and Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC
| | | | | | | | | |
Collapse
|
48
|
Abstract
The red cell is a relatively abundant locus of both free radical generation and reaction. Erythrocytes have a high content of unsaturated membrane lipids, a rich oxygen supply and are densely packed with redox-active hemoglobin residues. In response, red cells have a highly evolved and well-integrated network of oxidant defense mechanisms that lend an ability to withstand oxidative stress. In the case of congenital hemoglobin mutations that underlie sickle cell disease, they become very susceptible to free radical-mediated injury by virtue of enhanced endogenous rates of production of reactive species and impairment of tissue free radical defense mechanisms. In sickle cell disease, a combination of these susceptibility factors are hypothesized to lead to an overall impairment of vascular function, in large part due to loss of "bioactive" nitric oxide via the free radical-mediated consumption of this vasoactive molecule.
Collapse
Affiliation(s)
- M Aslan
- Department of Anesthesiology, University of Alabama at Birmingham 35233, USA
| | | | | |
Collapse
|
49
|
|
50
|
Affiliation(s)
- M T Gladwin
- Department of Critical Care Medicine, NIDDK National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|