1
|
Khan F, Ali H, Musharraf SG. Tenofovir disoproxil fumarate induces fetal hemoglobin production in K562 cells and β-YAC transgenic mice: A therapeutic approach for γ-globin induction. Exp Cell Res 2020; 394:112168. [PMID: 32653411 DOI: 10.1016/j.yexcr.2020.112168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023]
Abstract
Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating β-hemoglobinopathies like β-thalassemia and sickle cell anemia by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant nonresponders and toxicity limit its clinical usefulness. This study relates preclinical investigation of Tenofovir disoproxil fumarate (TDF) as a potential HbF inducing agent, using human erythroleukemia cell line and a β-YAC mouse model. Erythroid induction of K562 cells was studied by the benzidine/H2O2 reaction, total hemoglobin production was estimated by plasma hemoglobin assay kit, and γ-globin gene expression by RT-qPCR, whereas, fetal hemoglobin production was estimated by flow cytometry and immunofluorescence microscopy. We observed significantly increased γ- globin gene transcription and HbF expression mediated by TDF in K562 cells. Subsequent treatment of β-YAC transgenic mice with TDF confirmed HbF induction in vivo through an increase in γ-globin gene expression and in the percentage of HbF positive red blood cells. Moreover, TDF showed no cytotoxic effect at HbF inducing concentrations. These data support the potential development of TDF for the treatment of hematological disorders, including β-thalassemia and sickle cell anemia.
Collapse
Affiliation(s)
- Faisal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Hamad Ali
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Syed Ghulam Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
2
|
Miller AC, Rivas R, McMahon R, Miller K, Tesoro L, Villa V, Yanushkevich D, Lison P. Radiation protection and mitigation potential of phenylbutyrate: delivered via oral administration. Int J Radiat Biol 2017; 93:907-919. [DOI: 10.1080/09553002.2017.1350301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alexandra C. Miller
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
- Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Rafael Rivas
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Robert McMahon
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Karvelisse Miller
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Leonard Tesoro
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Vilmar Villa
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Daminik Yanushkevich
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| | - Paul Lison
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA
| |
Collapse
|
3
|
Boosalis MS, Sangerman JI, White GL, Wolf RF, Shen L, Dai Y, White E, Makala LH, Li B, Pace BS, Nouraie M, Faller DV, Perrine SP. Novel Inducers of Fetal Globin Identified through High Throughput Screening (HTS) Are Active In Vivo in Anemic Baboons and Transgenic Mice. PLoS One 2015; 10:e0144660. [PMID: 26713848 PMCID: PMC4694699 DOI: 10.1371/journal.pone.0144660] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/20/2015] [Indexed: 02/07/2023] Open
Abstract
High-level fetal (γ) globin expression ameliorates clinical severity of the beta (β) hemoglobinopathies, and safe, orally-bioavailable γ-globin inducing agents would benefit many patients. We adapted a LCR-γ-globin promoter-GFP reporter assay to a high-throughput robotic system to evaluate five diverse chemical libraries for this activity. Multiple structurally- and functionally-diverse compounds were identified which activate the γ-globin gene promoter at nanomolar concentrations, including some therapeutics approved for other conditions. Three candidates with established safety profiles were further evaluated in erythroid progenitors, anemic baboons and transgenic mice, with significant induction of γ-globin expression observed in vivo. A lead candidate, Benserazide, emerged which demonstrated > 20-fold induction of γ-globin mRNA expression in anemic baboons and increased F-cell proportions by 3.5-fold in transgenic mice. Benserazide has been used chronically to inhibit amino acid decarboxylase to enhance plasma levels of L-dopa. These studies confirm the utility of high-throughput screening and identify previously unrecognized fetal globin inducing candidates which can be developed expediently for treatment of hemoglobinopathies.
Collapse
Affiliation(s)
- Michael S. Boosalis
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jose I. Sangerman
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gary L. White
- Department of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Roman F. Wolf
- Department of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Ling Shen
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Yan Dai
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Emily White
- Department of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Levi H. Makala
- Department of Pediatrics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Biaoru Li
- Department of Pediatrics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Betty S. Pace
- Department of Pediatrics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Mehdi Nouraie
- Department of Medicine, Center for Sickle Cell Disease, Howard University, Washington, DC, United States of America
| | - Douglas V. Faller
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Phoenicia BioSciences, Weston, Massachusetts, United States of America
| | - Susan P. Perrine
- Cancer Center and Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Phoenicia BioSciences, Weston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
4
|
Kaddam L, FdleAlmula I, Eisawi OA, Abdelrazig HA, Elnimeiri M, Lang F, Saeed AM. Gum Arabic as fetal hemoglobin inducing agent in sickle cell anemia; in vivo study. BMC HEMATOLOGY 2015; 15:19. [PMID: 26719803 PMCID: PMC4696138 DOI: 10.1186/s12878-015-0040-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 12/10/2015] [Indexed: 11/29/2022]
Abstract
Background High levels of fetal haemoglobin (HbF) decrease sickle cell anaemia (SCA) severity and leads to improved survival. According to in vivo and in vitro studies, butyrate increases HbF production. Its utilization in clinical practice is hampered, however, by its short half-life. Serum butyrate concentrations could be enhanced by colonic bacterial fermentation of Gum Arabic (GA), edible, dried, gummy exudates from Acacia Senegal tree. We hypothesized that regular intake of GA increases serum butyrate levels, thus inducing HbF production and ameliorating symptoms of sickle cell anemia. Methods Fourty seven patients (5–42 years) carrying hemoglobin SS were recruited from April 2014 to January 2015. Patients received 30 g/day GA for 12 weeks. HbF, blood count and erythropoietin level were measured. The main outcome of interest was the level of HbF after 12 weeks. The secondary outcomes were improvement in clinical and laboratory results. The study was ethically approved by Alneelain University IRB. Results The study revealed significant increase in HbF level P.V0.000 [95 % CI, 0.43–1.02], MCV P.V:000 [95 % CI, 2.312–6.058] and Hematocrit level P.V:0.026 [95 % CI, 0.124–1.902]. No significant difference was encountered in platelets count P.V: 0.346 [95 % CI,−25.76–71.94], and WBCs count P.V:0.194 [95 % CI,−8.035–1.68]. Thirty seven percent of patients experienced minor side effects which resolved within a week. Conclusion These findings reveal a novel effect of GA, which may be used to foster fetal hemoglobin production. Trial registration ClinicalTrials.gov Identifier: NCT02467257. Registered 3rd June 2015. Electronic supplementary material The online version of this article (doi:10.1186/s12878-015-0040-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lamis Kaddam
- Department of Physiology Faculty of Medicine, Alneelain University, P.O. Box: 11121, Khartoum, 12702 Sudan
| | - Imad FdleAlmula
- Alneelain Research Centre Faculty of Medicine, Alneelain University, Khartoum, Sudan
| | - Omer Ali Eisawi
- Department of Hematology, Military Hospital, Khartoum, Sudan
| | | | - Mustafa Elnimeiri
- Department of Community Medicine Faculty of Medicine, Alneelain University, Khartoum, Sudan
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Amal M Saeed
- Department of Physiology Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
5
|
Loiselle K, Lee JL, Szulczewski L, Drake S, Crosby LE, Pai ALH. Systematic and Meta-Analytic Review: Medication Adherence Among Pediatric Patients With Sickle Cell Disease. J Pediatr Psychol 2015; 41:406-18. [PMID: 26384715 DOI: 10.1093/jpepsy/jsv084] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 08/13/2015] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To provide a comprehensive summary (systematic review) of medication adherence rates by assessment method and medication type for pediatric patients with sickle cell disease (SCD), as well as identify important correlates for future research. METHODS Articles assessing medication adherence and published between 1982 and February 2015 (n = 49) were identified using electronic databases. A meta-analysis of 14 studies examining demographic, medical, and psychosocial factors and medication adherence was conducted. RESULTS Adherence rates ranged from 12% to 100% across all medications. Approximately 30% of studies reported associations between adherence and key demographic, medical, and psychosocial correlates. Mean effect sizes were small to moderate (r = .02-.53). CONCLUSIONS The wide range of adherence rates reported in the literature may be because of, in part, the use of variable assessment strategies. Future studies examining pediatric SCD adherence should incorporate key correlates with the goal of replication.
Collapse
Affiliation(s)
- Kristin Loiselle
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Jennifer L Lee
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Lauren Szulczewski
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Sarah Drake
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Lori E Crosby
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Ahna L H Pai
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| |
Collapse
|
6
|
The LSD1 inhibitor RN-1 induces fetal hemoglobin synthesis and reduces disease pathology in sickle cell mice. Blood 2015; 126:386-96. [PMID: 26031919 DOI: 10.1182/blood-2015-02-626259] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/30/2022] Open
Abstract
Inhibition of lysine-specific demethylase 1 (LSD1) has been shown to induce fetal hemoglobin (HbF) levels in cultured human erythroid cells in vitro. Here we report the in vivo effects of LSD1 inactivation by a selective and more potent inhibitor, RN-1, in a sickle cell disease (SCD) mouse model. Compared with untreated animals, RN-1 administration leads to induced HbF synthesis and to increased frequencies of HbF-positive cells and mature erythrocytes, as well as fewer reticulocytes and sickle cells, in the peripheral blood of treated SCD mice. In keeping with these observations, histologic analyses of the liver and spleen of treated SCD mice verified that they do not exhibit the necrotic lesions that are usually associated with SCD. These data indicate that RN-1 can effectively induce HbF levels in red blood cells and reduce disease pathology in SCD mice, and may therefore offer new therapeutic possibilities for treating SCD.
Collapse
|
7
|
What are the most promising emerging therapies for sickle cell disease? Future Med Chem 2014; 6:979-82. [DOI: 10.4155/fmc.14.55] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
8
|
Kim SW, Hooker JM, Otto N, Win K, Muench L, Shea C, Carter P, King P, Reid AE, Volkow ND, Fowler JS. Whole-body pharmacokinetics of HDAC inhibitor drugs, butyric acid, valproic acid and 4-phenylbutyric acid measured with carbon-11 labeled analogs by PET. Nucl Med Biol 2013; 40:912-8. [PMID: 23906667 DOI: 10.1016/j.nucmedbio.2013.06.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/09/2013] [Accepted: 06/16/2013] [Indexed: 12/16/2022]
Abstract
The fatty acids, n-butyric acid (BA), 4-phenylbutyric acid (PBA) and valproic acid (VPA, 2-propylpentanoic acid) have been used for many years in the treatment of a variety of CNS and peripheral organ diseases including cancer. New information that these drugs alter epigenetic processes through their inhibition of histone deacetylases (HDACs) has renewed interest in their biodistribution and pharmacokinetics and the relationship of these properties to their therapeutic and side effect profiles. In order to determine the pharmacokinetics and biodistribution of these drugs in primates, we synthesized their carbon-11 labeled analogues and performed dynamic positron emission tomography (PET) in six female baboons over 90 min. The carbon-11 labeled carboxylic acids were prepared by using (11)CO2 and the appropriate Grignard reagents. [(11)C]BA was metabolized rapidly (only 20% of the total carbon-11 in plasma was parent compound at 5 min post injection) whereas for VPA and PBA 98% and 85% of the radioactivity were the unmetabolized compound at 30 min after their administration respectively. The brain uptake of all three carboxylic acids was very low (<0.006%ID/cc, BA>VPA>PBA), which is consistent with the need for very high doses for therapeutic efficacy. Most of the radioactivity was excreted through the kidneys and accumulated in the bladder. However, the organ biodistribution between the drugs differed. [(11)C]BA showed relatively high uptake in spleen and pancreas whereas [(11)C]PBA showed high uptake in liver and heart. Notably, [(11)C]VPA showed exceptionally high heart uptake possibly due to its involvement in lipid metabolism. The unique biodistribution of each of these drugs may be of relevance in understanding their therapeutic and side effect profile including their teratogenic effects.
Collapse
Affiliation(s)
- Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Upton, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Kutlar A, Ataga K, Reid M, Vichinsky EP, Neumayr L, Blair-Britt L, Labotka R, Glass J, Keefer JR, Wargin WA, Berenson R, Perrine SP. A phase 1/2 trial of HQK-1001, an oral fetal globin inducer, in sickle cell disease. Am J Hematol 2012; 87:1017-21. [PMID: 22887019 DOI: 10.1002/ajh.23306] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 12/18/2022]
Abstract
Therapeutics which reduce the pathology in sickle cell syndromes are needed, particularly noncytotoxic therapeutics. Fetal hemoglobin (HbF, α(2) γ(2) ) is established as a major regulator of disease severity; increased HbF levels correlate with milder clinical courses and improved survival. Accordingly, sodium dimethylbutyrate (HQK-1001), an orally-bioavailable, promoter-targeted fetal globin gene-inducing agent, was evaluated in a randomized, blinded, dose-ranging Phase I/II trial in 24 adult patients with HbSS or S/β thalassemia, to determine safety and tolerability of three escalating dose levels. The study therapeutic was administered once daily for two 6-week cycles, with a two-week interim dose holiday. Twenty-one patients completed the study. Five patients received study drug at 10 or 20 mg/kg doses, seven patients received study drug at 30 mg/kg/dose, and 4 patients received placebo. HQK-1001 was well-tolerated with no unexpected drug-related adverse events; a dose-limiting toxicity was not identified. Plasma drug levels were sustained above targeted levels for 24 hr. Increases in HbF above baseline were observed particularly with 30 mg/kg/day doses; in five of seven treated patients, a mean absolute increase in HbF of 0.2 g/dl and a mean increase in total hemoglobin (Hgb) of 0.83 g/dl above baseline were observed, whereas no increases occurred in placebo-treated controls. These findings of favorable PK profiles, tolerability, early rises in HbF, and total Hgb indicate that trials of longer duration appear warranted to more definitively evaluate the therapeutic potential of HQK-1001 in sickle cell disease.
Collapse
Affiliation(s)
- Abdullah Kutlar
- Department of Medicine, Sickle Cell Center, Medical College of Georgia, Augusta, GA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Miller AC, Cohen S, Stewart M, Rivas R, Lison P. Radioprotection by the histone deacetylase inhibitor phenylbutyrate. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2011; 50:585-596. [PMID: 21892632 DOI: 10.1007/s00411-011-0384-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 08/15/2011] [Indexed: 05/31/2023]
Abstract
The histone deacetylase inhibitor (HDAC), phenylbutyrate (PB), is a novel anti-tumor agent. Studies have demonstrated that HDAC inhibitors can suppress cutaneous radiation syndrome and stimulate hematopoiesis. The objective of this study was to test the ability of PB treatment to protect against acute gamma-radiation-induced lethality in the DBA/2 mouse model. A 30-day radiation lethality study was used to assess radioprotective capability of PB. Mechanisms were evaluated using western blots, flow cytometry, and the single-cell gel electrophoresis assay. Western blot studies showed that PB treatment acetylated histones in vivo. For radiation protection studies, prophylactic administration of PB (24 h preradiation; 1-50 mg/kg) provided radioprotection against gamma radiation (8-9.5 Gy) and PB demonstrated a DRF of 1.31 (P = 0.001; 95% confidence interval: 1.27, 1.36). When PB (10 mg/kg) was administered post-radiation (4 h), it also provided significant radioprotection at 8.0 Gy radiation (P = 0.022). PB treatment before radiation was associated with significant elevations in neutrophils and platelets following radiation. Results from single-cell gel electrophoresis of peripheral blood leukocytes demonstrated that PB treatment before radiation can attenuate DNA damage and inhibit radiation-induced apoptosis. These results indicate that an HDAC inhibitor like PB has potential as a radiation protector and that mechanisms of action include attenuation of DNA damage and inhibition of apoptosis.
Collapse
Affiliation(s)
- Alexandra C Miller
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University, Bethesda, MD 20889-5603, USA.
| | | | | | | | | |
Collapse
|
11
|
Boosalis MS, Castaneda SA, Trudel M, Mabaera R, White GL, Lowrey CH, Emery DW, Mpollo MSEM, Shen L, Wargin WA, Bohacek R, Faller DV, Perrine SP. Novel therapeutic candidates, identified by molecular modeling, induce γ-globin gene expression in vivo. Blood Cells Mol Dis 2011; 47:107-16. [PMID: 21641240 PMCID: PMC3148318 DOI: 10.1016/j.bcmd.2011.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 11/25/2022]
Abstract
The β-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the β-globin chain of hemoglobin A (α(2)β(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and β-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.
Collapse
Affiliation(s)
- Michael S. Boosalis
- Cancer Center and Hemoglobinopathy-Thalassemia Research Unit, Boston University School of Medicine, Boston, MA, USA
| | - Serguei A. Castaneda
- Cancer Center and Hemoglobinopathy-Thalassemia Research Unit, Boston University School of Medicine, Boston, MA, USA
| | - Marie Trudel
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montreal, Universite de Montreal, Montreal, Quebec, Canada, USA
| | - Rodwell Mabaera
- Departments of Medicine, Pharmacology, and Toxicology, Dartmouth Medical School, Hanover, NH, USA
| | - Gary L. White
- Department of Comparative Medicine Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Christopher H. Lowrey
- Departments of Medicine, Pharmacology, and Toxicology, Dartmouth Medical School, Hanover, NH, USA
| | - David W. Emery
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marthe-Sandrine Eiymo Mwa Mpollo
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montreal, Universite de Montreal, Montreal, Quebec, Canada, USA
| | - Ling Shen
- Cancer Center and Hemoglobinopathy-Thalassemia Research Unit, Boston University School of Medicine, Boston, MA, USA
| | | | | | - Douglas V. Faller
- Cancer Center and Hemoglobinopathy-Thalassemia Research Unit, Boston University School of Medicine, Boston, MA, USA
| | - Susan P. Perrine
- Cancer Center and Hemoglobinopathy-Thalassemia Research Unit, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Perrine SP, Wargin WA, Boosalis MS, Wallis WJ, Case S, Keefer JR, Faller DV, Welch WC, Berenson RJ. Evaluation of safety and pharmacokinetics of sodium 2,2 dimethylbutyrate, a novel short chain fatty acid derivative, in a phase 1, double-blind, placebo-controlled, single-dose, and repeat-dose studies in healthy volunteers. J Clin Pharmacol 2011; 51:1186-94. [PMID: 21422239 DOI: 10.1177/0091270010379810] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pharmacologic induction of fetal globin synthesis is an accepted therapeutic strategy for treatment of the beta hemoglobinopathies and thalassemias, as even small increases in hemoglobin F (HbF) levels reduce clinical severity in sickle cell disease (SCD) and reduce anemia in beta thalassemia. Prior generation short chain fatty acid therapeutics, arginine butyrate (AB), and phenylbutyrate, increased fetal and total hemoglobin levels in patients, but were limited by high doses or intravenous (IV) infusion. A fetal globin-inducing therapeutic with convenient oral dosing would be an advance for these classic molecular diseases. Healthy adult human subjects were treated with a novel short chain fatty acids (SCFA) derivative, sodium 2,2 dimethylbutyrate (SDMB), or placebo, with 1 of 4 single dose levels (2, 5, 10, and 20 mg/kg) or daily doses (5, 10, or 15 mg/kg) over 14 days, and monitored for adverse clinical and laboratory events, drug levels, reticulocytes, and HbF assays. SDMB was well-tolerated with no clinically significant adverse events related to study medication. The terminal half-life ranged from 9 to 15 hours. Increases in mean absolute reticulocytes were observed at all dose levels in the 14-day study. The favorable pharmacokinetics (PK) profiles and safety findings indicate that SDMB warrants further investigation for treatment of anemic subjects with beta hemoglobinopathies.
Collapse
|
13
|
Basseri S, Lhoták S, Sharma AM, Austin RC. The chemical chaperone 4-phenylbutyrate inhibits adipogenesis by modulating the unfolded protein response. J Lipid Res 2010; 50:2486-501. [PMID: 19461119 DOI: 10.1194/jlr.m900216-jlr200] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent studies have shown a link between obesity and endoplasmic reticulum (ER) stress. Perturbations in ER homeostasis cause ER stress and activation of the unfolded protein response (UPR). Adipocyte differentiation contributes to weight gain, and we have shown that markers of ER stress/UPR activation, including GRP78, phospho-eIF2, and spliced XBP1, are upregulated during adipogenesis. Given these findings, the objective of this study was to determine whether attenuation of UPR activation by the chemical chaperone 4-phenylbutyrate (4-PBA) inhibits adipogenesis. Exposure of 3T3-L1 preadipocytes to 4-PBA in the presence of differentiation media decreased expression of ER stress markers. Concomitant with the suppression of UPR activation, 4-PBA resulted in attenuation of adipogenesis as measured by lipid accumulation and adiponectin secretion. Consistent with these in vitro findings, female C57BL/6 mice fed a high-fat diet supplemented with 4-PBA showed a significant reduction in weight gain and had reduced fat pad mass, as compared with the high-fat diet alone group. Furthermore, 4-PBA supplementation decreased GRP78 expression in the adipose tissue and lowered plasma triglyceride, glucose, leptin, and adiponectin levels without altering food intake. Taken together, these results suggest that UPR activation contributes to adipogenesis and that blocking its activation with 4-PBA prevents adipocyte differentiation and weight gain in mice.
Collapse
Affiliation(s)
- Sana Basseri
- Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton and the Henderson Research Centre, Hamilton, Ontario, L8N 4A6, Canada
| | | | | | | |
Collapse
|
14
|
Abstract
Sickle cell anemia is one of the most common autosomal recessive diseases in the world. Patients with sickle cell anemia have variable penetrance and it is hard to predict the risk and timing of complications. It is characterized by a point mutation in the beta-globin gene (GAG --> GTG) and the production of hemoglobin S. The latter leads to decreased deformability of the red blood cells (RBCs) that adhere to endothelia cells culminating in vascular occlusion and its sequelae of tissue ischemia and organ damage. Moreover, sickled RBCs undergo intravascular hemolysis and accelerated erythropoesis. The hallmarks of this disease are shortened RBC survival and vaso-occlusive crises. For the past ten years, the pathophysiology of this disease has been better elucidated and has led to significant improvements in the standard of care. Vaso-occlusion is now understood to be a complex event that involves abnormal interactions between RBCs, leukocytes, endothelial cells and the coagulation pathways. The field of translational research in sickle cell anemia has expanded greatly and has led to new clinical trials with new therapeutic agents and strategies. In this paper, we review the drugs that are now being investigated in the treatment of sickle cell anemia.
Collapse
|
15
|
Abstract
Hydroxyurea therapy offers promise for ameliorating the clinical course of children with sickle cell disease (SCD). Hydroxyurea is a prototypic therapeutic option; it can be administered with minimal side effects, has a relatively wide therapeutic window, and has mechanisms of action that address pathophysiologic pathways of sickling, vaso-occlusion, hemolysis, and organ damage. There are limited data regarding hydroxyurea's ability to prevent or diminish organ dysfunction, and the long-term risks of hydroxyurea therapy remain incompletely defined. Although clinical trials are underway to address long-term issues, hydroxyurea remains an effective but underutilized therapy for SCD.
Collapse
Affiliation(s)
- Matthew M Heeney
- Department of Pediatrics, Harvard Medical School, Children's Hospital Boston, 300 Longwood Avenue, Boston, MA 02115, USA.
| | | |
Collapse
|
16
|
Yao X, Kodeboyina S, Liu L, Dzandu J, Sangerman J, Ofori-Acquah SF, Pace BS. Role of STAT3 and GATA-1 interactions in gamma-globin gene expression. Exp Hematol 2009; 37:889-900. [PMID: 19447160 PMCID: PMC2746970 DOI: 10.1016/j.exphem.2009.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2008] [Revised: 04/06/2009] [Accepted: 05/08/2009] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We previously demonstrated a silencing role for signal transducers and activators of transcription 3 (STAT3) in gamma-globin gene regulation in primary erythroid cells. Recently, GATA-1, a key transcription factor involved in hematopoietic cell development, was shown to directly inhibit STAT3 activity in vivo. Therefore, we completed studies to determine if interactions between these two factors influence gamma-globin gene expression. MATERIALS AND METHODS Chromatin immunoprecipitation assay was used to ascertain in vivo protein binding at the gamma-globin 5' untranslated region (5'UTR); protein-protein interactions were examined by coimmunoprecipitation analysis. In vitro protein-DNA binding were completed using surface plasmon resonance and electrophoretic mobility shift assay. Activity of a luciferase gamma-globin promoter reporter and levels of gamma-globin messenger RNA and fetal hemoglobin in stable K562 cell lines overexpressing STAT3 and GATA-1, were used to determine the influence of the STAT3/GATA-1 interaction on gamma-globin gene expression. RESULTS We observed interaction between STAT3 and GATA-1 in K562 and mouse erythroleukemia cells in vivo at the gamma-globin 5'UTR by chromatin immunoprecipitation assay. Electrophoretic mobility shift assay performed with a 41-base pair gamma-globin DNA probe (gamma41) demonstrated the presence of STAT3 and GATA-1 proteins in complexes assembled at the gamma-globin 5'UTR. A consensus STAT3 DNA probe inhibited GATA-1-binding in a concentration-dependent manner, and the converse was also true. Enforced STAT3 expression augmented its binding at the gamma-globin 5'UTR in vivo and silenced gamma-promoter-driven luciferase activity. Stable enforced STAT3 expression in K562 cells reduced endogenous gamma-globin messenger RNA level. This effect was reversed by GATA-1. CONCLUSION These data provide evidence that GATA-1 can reverse STAT3-mediated gamma-globin gene silencing in erythroid cells.
Collapse
Affiliation(s)
- Xiao Yao
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX
| | - Sirisha Kodeboyina
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX
| | - Li Liu
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX
| | - James Dzandu
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX
| | - Jose Sangerman
- Yale University, Department of Pediatrics, New Haven, CT
| | | | - Betty S. Pace
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, TX
| |
Collapse
|
17
|
Abstract
In the last decade, the care of patients with sickle cell disease (SCD) has undergone important advances with better understanding of disease pathophysiology and improvement in standards of care, especially among paediatric patients. Although many new drugs are currently being investigated and are at different stages of development, the pace of drug discovery and utilization has been slow and suboptimal. Hydroxycarbamide (hydroxyurea) has been investigated and utilized for at least two decades. Hydroxycarbamide's efficacy has been demonstrated, albeit with different levels of evidence, in paediatric and adult populations, and yet clinician and patient acceptance and use have been far from ideal. In this review we discuss the current usage of hydroxycarbamide and its possible future indications in SCD, as well as the use of new compounds that have very different mechanisms of action, which may prove safe and efficacious when used alone or in combination in patients with SCD.
Collapse
Affiliation(s)
- Jane Hankins
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA.
| | | |
Collapse
|
18
|
Abstract
While supportive care remains the best option for most well children with sickle cell disease (SCD), increasing awareness of early signs of chronic organ damage in childhood has focused attention on therapy which modulates the natural history of the disease. Since cure by stem cell transplantation is only feasible for a minority and gene therapy remains developmental, pharmacological modification by Haemoglobin F (HbF)-inducers, is the most widely used approach in SCD. Currently, the only HbF modulator with a clear place in the management of childhood SCD is hydroxycarbamide for which the main indications are frequent painful crises and recurrent acute chest syndrome. In the majority of SCD children treated with hydroxycarbamide there is clear evidence of clinical benefit and the drug is well tolerated. The main disadvantages are the need for frequent monitoring and uncertainity about long-term risks of carcinogenicity and impaired fertility, although these risks appear to be very low. The role of hydroxycarbamide in sickle-associated central nervous system disease remains to be established. Decitabine and butyrate derivatives show some promise although robust data in children with SCD are lacking. A number of other drugs are currently under investigation for their effects on HbF production including thalidomide and lenolidamide.
Collapse
Affiliation(s)
- Sara Trompeter
- Department of Haematology, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | | |
Collapse
|
19
|
Fetal hemoglobin chemical inducers for treatment of hemoglobinopathies. Ann Hematol 2008; 88:505-28. [PMID: 19011856 DOI: 10.1007/s00277-008-0637-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 10/27/2008] [Indexed: 12/29/2022]
Abstract
The switch from fetal ((G)gamma and (A)gamma) to adult (beta and delta) globin gene expression occurs at birth, leading to the gradual replacement of HbF with HbA. Genetic regulation of this switch has been studied for decades, and the molecular mechanisms underlying this developmental change in gene expression have been in part elucidated. The understanding of the developmental regulation of gamma-globin gene expression was paralleled by the identification of a series of chemical compounds able to reactivate HbF synthesis in vitro and in vivo in adult erythroid cells. Reactivation of HbF expression is an important therapeutic option in patients with hemoglobin disorders, such as sickle cell anemia and beta-thalassemia. These HbF inducers can be grouped in several classes based on their chemical structures and mechanisms of action. Clinical studies with some of these agents have shown that they were effective, in a part of patients, in ameliorating the clinical condition. The increase in HbF in response to these drugs varies among patients with beta-thalassemia and sickle cell disease due to individual genetic determinants.
Collapse
|
20
|
|
21
|
Perrine SP. Fetal globin stimulant therapies in the beta-hemoglobinopathies: principles and current potential. Pediatr Ann 2008; 37:339-46. [PMID: 18543545 DOI: 10.3928/00904481-20080501-10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
For the majority of children with beta- hemoglobinopathies and -thalassemias who do not have a transplant donor, survival is shortened and morbidity is high. Hydroxyurea, EPO preparations, sodium phenylbutyrate, arginine butyrate, and 5-azacytidine/decitabine have shown efficacy in approximately 40% to 70% of sickle cell and beta-thalassemia patients. Many responses, although significant, were not completely ameliorating of symptoms or pathology, and trials of new agents with dual actions, or drug combinations, are needed. Ideally, limiting chemotherapeutic exposure is desirable for long-term treatment of children, and an oral therapeutic at tolerable doses is necessary for practical use. A new oral therapeutic candidate that induces fetal hemoglobin production and also stimulates erythropoiesis is entering clinical evaluation. Use of agents that should have additive or synergistic effects in combination, such as EPO and hydroxyurea or a short-chain fatty acid derivative (SCFAD), offer better therapeutic potential than hydroxyurea alone. Childhood is an optimal time to introduce such therapies, particularly the non-mutagenic SCFADs, while the erythroid marrow reserve is preserved and before organ damage has become widespread. A challenge for successful application of these therapies is to define patient subsets that are most likely to respond to a particular agent, or which require combination therapies, and to develop optimal dose regimens in thalassemias with rapid erythroid apoptosis. Development of this therapeutic avenue will require close collaboration among treating and academic physicians, families and patients, funding agencies, and researchers.
Collapse
Affiliation(s)
- Susan P Perrine
- Hemoglobinopathy Thalassemia Research Unit, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
22
|
Abstract
Hydroxyurea therapy offers promise for ameliorating the clinical course of children with sickle cell disease (SCD). Hydroxyurea is a prototypic therapeutic option; it can be administered with minimal side effects, has a relatively wide therapeutic window, and has mechanisms of action that address pathophysiologic pathways of sickling, vaso-occlusion, hemolysis, and organ damage. There are limited data regarding hydroxyurea's ability to prevent or diminish organ dysfunction, and the long-term risks of hydroxyurea therapy remain incompletely defined. Although clinical trials are underway to address long-term issues, hydroxyurea remains an effective but underutilized therapy for SCD.
Collapse
|
23
|
Burkitt K, Ljungman M. Phenylbutyrate interferes with the Fanconi anemia and BRCA pathway and sensitizes head and neck cancer cells to cisplatin. Mol Cancer 2008; 7:24. [PMID: 18325101 PMCID: PMC2276233 DOI: 10.1186/1476-4598-7-24] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Accepted: 03/06/2008] [Indexed: 12/14/2022] Open
Abstract
Background Cisplatin has been widely used to treat head and neck cancer. One of the clinical limitations with this treatment, however, is that tumors that are initially responsive to cisplatin later acquire resistance. We have recently shown that a subset of head and neck cancer cell lines has a defective Fanconi anemia DNA damage response pathway and this defect correlates to cisplatin sensitivity. We have also shown that the histone deacetylase inhibitor phenylbutyrate sensitize human cells to cisplatin. In this study we explored whether phenylbutyrate may sensitize head and neck cancer cells by interfering with the Fanconi anemia pathway. Results We found that the phenylbutyrate sensitizes head and neck cancer cell lines to cisplatin. This sensitization by phenylbutyrate correlated to a significant decrease in the formation of cisplatin-induced FANCD2 nuclear foci, which is a functional read out of the Fanconi anemia and BRCA (FA/BRCA) pathway. This abrogation of the FA/BRCA pathway by phenylbutyrate was not due to loss of FANCD2 monoubiquitylation but rather correlated to a phenylbutyrate-mediated reduction in the expression of the BRCA1 protein. Furthermore, we found that cancer cells defective in the FA pathway were also sensitized to cisplatin by phenylbutyrate suggesting that phenylbutyrate targets additional pathways. Conclusion The results from this study suggest that phenylbutyrate may have therapeutic utility as a cisplatin sensitizer in head and neck cancer by inhibiting the FA/BRCA pathway through the down regulation of BRCA1 as well as by an FA/BRCA-independent mechanism.
Collapse
Affiliation(s)
- Kyunghee Burkitt
- Department of Radiation Oncology, Division of Radiation Cancer Biology, University of Michigan Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
24
|
Hines P, Dover GJ, Resar LMS. Pulsed-dosing with oral sodium phenylbutyrate increases hemoglobin F in a patient with sickle cell anemia. Pediatr Blood Cancer 2008; 50:357-9. [PMID: 17253639 DOI: 10.1002/pbc.21104] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Increasing hemoglobin F (HbF) appears to be beneficial for patients with sickle cell anemia. We previously demonstrated that daily, oral sodium phenylbutyrate (OSPB) induces HbF synthesis in pediatric and adult patients with hemoglobin SS (HbSS). The high doses and need for daily therapy, however, have limited its use. Here, we report a patient treated with pulsed-dosing of OSPB for over 3 years. This patient developed a modest, but sustained elevation in HbF over the course of therapy without side effects. Although larger studies are needed, this case demonstrates that pulsed-dosing with OSPB enhances HbF synthesis.
Collapse
Affiliation(s)
- Patrick Hines
- Hematology Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
25
|
Lopez CA, Feng FY, Herman JM, Nyati MK, Lawrence TS, Ljungman M. Phenylbutyrate sensitizes human glioblastoma cells lacking wild-type p53 function to ionizing radiation. Int J Radiat Oncol Biol Phys 2007; 69:214-20. [PMID: 17707275 DOI: 10.1016/j.ijrobp.2007.04.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Revised: 04/15/2007] [Accepted: 04/18/2007] [Indexed: 01/25/2023]
Abstract
PURPOSE Histone deacetylase (HDAC) inhibitors induce growth arrest, differentiation, and apoptosis in cancer cells. Phenylbutyrate (PB) is a HDAC inhibitor used clinically for treatment of urea cycle disorders. Because of its low cytotoxicity, cerebrospinal fluid penetration, and high oral bioavailability, we investigated PB as a potential radiation sensitizer in human glioblastoma cell lines. METHODS AND MATERIALS Four glioblastoma cell lines were selected for this study. Phenylbutyrate was used at a concentration of 2 mM, which is achievable in humans. Western blots were used to assess levels of acetylated histone H3 in tumor cells after treatment with PB. Flow cytometry was used for cell cycle analysis. Clonogenic assays were performed to assess the effect of PB on radiation sensitivity. We used shRNA against p53 to study the role of p53 in radiosensitization. RESULTS Treatment with PB alone resulted in hyperacetylation of histones, confirmed by Western blot analysis. The PB alone resulted in cytostatic effects in three cell lines. There was no evidence of G(1) arrest, increase in sub-G(1) fraction or p21 protein induction. Clonogenic assays showed radiosensitization in two lines harboring p53 mutations, with enhancement ratios (+/- SE) of 1.5 (+/- 0.2) and 1.3 (+/- 0.1), respectively. There was no radiopotentiating effect in two cell lines with wild-type p53, but knockdown of wild-type p53 resulted in radiosensitization by PB. CONCLUSIONS Phenylbutyrate can produce p21-independent cytostasis, and enhances radiation sensitivity in p53 mutant human glioblastoma cells in vitro. This suggests the potential application of combined PB and radiotherapy in glioblastoma harboring mutant p53.
Collapse
Affiliation(s)
- Carlos A Lopez
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109-0010, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Odièvre MH, Brun M, Krishnamoorthy R, Lapouméroulie C, Elion J. Sodium phenyl butyrate downregulates endothelin-1 expression in cultured human endothelial cells: relevance to sickle-cell disease. Am J Hematol 2007; 82:357-62. [PMID: 17373676 DOI: 10.1002/ajh.20709] [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: 11/07/2022]
Abstract
As hydroxyurea (HU), sodium phenyl butyrate (SPB) is known to induce fetal hemoglobin (HbF) expression and thus shows potentials for sickle-cell disease (SCD) treatment. More recently, few studies suggested that endothelial cells (ECs), a major pathophysiological actor of SCD, are also a target of SPB. Here, we show that SPB, as HU, reduces endothelin-1 mRNA expression and peptide release by human ECs in culture. SPB increases VCAM-1 and ICAM-1 mRNAs and soluble ICAM-1 release. Both drugs have a cumulative effect on ICAM-1 expression. We conclude that SPB, as HU, also affects the expression of molecules important to the pathophysiology of SCD, in addition to its effect on HbF. Its potential as an alternative or adjuvant drug in SCD treatment warrants further investigations.
Collapse
|
27
|
Kern RM, Yang Z, Kim PS, Grody WW, Iyer RK, Cederbaum SD. Arginase induction by sodium phenylbutyrate in mouse tissues and human cell lines. Mol Genet Metab 2007; 90:37-41. [PMID: 16935537 DOI: 10.1016/j.ymgme.2006.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Accepted: 07/06/2006] [Indexed: 11/19/2022]
Abstract
Hyperargininemia is a urea cycle disorder caused by mutations in the gene for arginase I (AI) resulting in elevated blood arginine and ammonia levels. Sodium phenylacetate and a precursor, sodium phenylbutyrate (NaPB) have been used to lower ammonia, conjugating glutamine to produce phenylacetylglutamine which is excreted in urine. The elevated arginine levels induce the second arginase (AII) in patient kidney and kidney tissue culture. It has been shown that NaPB increases expression of some target genes and we tested its effect on arginase induction. Eight 9-week old male mice fed on chow containing 7.5 g NaPB/kg rodent chow and drank water with 10 g NaPB/L, and four control mice had a normal diet. After one week all mice were sacrificed. The arginase specific activities for control and NaPB mice, respectively, were 38.2 and 59.4 U/mg in liver, 0.33 and 0.42 U/mg in kidney, and 0.29 and 1.19 U/mg in brain. Immunoprecipitation of arginase in each tissue with AI and AII antibodies showed the activity induced by NaPB is mostly AI. AII may also be induced in kidney. AI accounts for the fourfold increased activity in brain. In some cell lines, NaPB increased arginase activity up to fivefold depending on dose (1-5 mM) and exposure time (2-5 days); control and NaPB activities, respectively, are: erythroleukemia, HEL, 0.06 and 0.31 U/mg, and K562, 0.46 and 1.74 U/mg; embryonic kidney, HEK293, 1.98 and 3.58 U/mg; breast adenocarcinoma, MDA-MB-468, 1.11 and 4.06 U/mg; and prostate adenocarcinoma, PC-3, 0.55 and 3.20 U/mg. In MDA-MB-468 and HEK most, but not all, of the induced activity is AI. These studies suggest that NaPB may induce AI when used to treat urea cycle disorders. It is relatively less useful in AI deficiency, although it could have some effect in those patients with missense mutations.
Collapse
Affiliation(s)
- R M Kern
- The Mental Retardation Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
28
|
Fathallah H, Atweh GF. Induction of fetal hemoglobin in the treatment of sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2006:58-62. [PMID: 17124041 DOI: 10.1182/asheducation-2006.1.58] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
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 inhibits the polymerization of sickle hemoglobin and the resulting pathophysiology. Hydroxyurea, an inducer of HbF, has already been approved for the treatment of patients with moderate and/or severe SCD. Recent clinical trials with other pharmacological inducers of HbF, such as butyrate and decitabine, have shown considerable promise. In this chapter, we highlight the important clinical trials with pharmacological inducers of HbF, discuss their mechanisms of action and speculate about the future of this therapeutic approach in the treatment of patients with SCD.
Collapse
Affiliation(s)
- Hassana Fathallah
- Mount Sinai School of Medicine, Division of Hematology and Oncology, Box 1079, One Gustave L. Levy Place, New York, NY 10029, USA.
| | | |
Collapse
|
29
|
Rishikof DC, Ricupero DA, Liu H, Goldstein RH. Phenylbutyrate decreases type I collagen production in human lung fibroblasts. J Cell Biochem 2004; 91:740-8. [PMID: 14991765 DOI: 10.1002/jcb.10742] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibrotic lung diseases are characterized by excess extracellular matrix production, in particular type I collagen. Phenylbutyrate (PB) is a non-toxic pharmacological compound that functions as a weak histone deacetylase inhibitor. In hepatic stellate cells, the synthesis of type I collagen expression is decreased by inhibiting histone acetylation. Our studies examined the regulation of type I collagen by PB in human lung fibroblasts. We found that PB decreases basal and transforming growth factor-beta-stimulated alpha1(I) collagen mRNA and protein levels. Northern blot analyses demonstrated that PB decreases steady-state alpha1(I) collagen mRNA levels by 78% without significantly changing the stability of the mRNA transcript. PB stimulates cAMP production and increases the acetylation of histone H4, but does not affect the activity of two transforming growth factor-beta (TGF-beta)-responsive luciferase reporter constructs. These data suggest that PB regulates type I collagen expression in human lung fibroblasts by mechanisms that include cAMP production and histone acetylation. PB may have therapeutic use in fibrotic lung diseases.
Collapse
Affiliation(s)
- David C Rishikof
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | | | |
Collapse
|