Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 202(1): 173-183, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36960712

RESUMO

The pathophysiologic mechanism of sickle cell disease (SCD) involves polymerization of deoxygenated haemoglobin S (HbS), leading to red blood cell (RBC) sickling, decreased RBC deformability, microvascular obstruction, haemolysis, anaemia and downstream clinical complications. Pharmacological increase in the concentration of oxygenated HbS in RBCs has been shown to be a novel approach to inhibit HbS polymerization and reduce RBC sickling and haemolysis. We report that GBT021601, a small molecule that increases HbS-oxygen affinity, inhibits HbS polymerization and prevents RBC sickling in blood from patients with SCD. Moreover, in a murine model of SCD (SS mice), GBT021601 reduces RBC sickling, improves RBC deformability, prolongs RBC half-life and restores haemoglobin levels to the normal range, while improving oxygen delivery and increasing tolerance to severe hypoxia. Notably, oral dosing of GBT021601 in animals results in higher levels of Hb occupancy than voxelotor and suggests the feasibility of once-daily dosing in humans. In summary, GBT021601 improves RBC health and normalizes haemoglobin in SS mice, suggesting that it may be useful for the treatment of SCD. These data are being used as a foundation for clinical research and development of GBT021601.


Assuntos
Anemia Falciforme , Hemólise , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Oxigênio , Anemia Falciforme/tratamento farmacológico , Eritrócitos , Hemoglobinas , Hemoglobina Falciforme
2.
Am J Physiol Heart Circ Physiol ; 321(2): H400-H411, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34213392

RESUMO

Therapeutic agents that increase the Hb affinity for oxygen (O2) could, in theory, lead to decreased O2 release from Hb and impose a hypoxic risk to tissues. In this study, GBT1118, an allosteric modifier of Hb affinity for O2, was used to assess the impact of increasing Hb affinity for O2 on brain tissue oxygenation, blood pressure, heart rate, O2 delivery, and tolerance to hypoxia in Townes transgenic sickle cell disease (SCD) mice. Brain oxygenation and O2 delivery were studied during normoxia and severe hypoxic challenges. Chronic treatment with GBT1118 increased Hb affinity for O2, reducing the Po2 for 50% HbO2 saturation (P50) in SCD mice from 31 mmHg to 18 mmHg. This treatment significantly reduced anemia, increasing hematocrit by 33%, improved cardiac output (CO), and O2 delivery and extraction. Chronically increasing Hb affinity for O2 with GBT1118 preserved cortical O2 tension during normoxia, improved cortical O2 tension during hypoxia, and increased tolerance to severe hypoxia in SCD mice. Independent of hematological changes induced by chronic treatment, a single dose of GBT1118 significantly improved tolerance to hypoxia, highlighting the benefits of increasing Hb affinity for O2 and consequently reducing sickling of RBCs in blood during hypoxia in SCD.NEW & NOTEWORTHY Chronic pharmacologically increased hemoglobin affinity for oxygen in sickle cell disease mice alleviated hematological consequences of sickle cell disease, increasing RBC half-life, hematocrit, and hemoglobin concentration, while also decreasing reticulocyte count. Additionally, chronically increased hemoglobin affinity for oxygen significantly improved survival as well as cortical tissue oxygenation in sickle cell disease mice during hypoxia, suggesting that oxygen delivery and utilization is improved by increased hemoglobin affinity for oxygen.


Assuntos
Anemia Falciforme/metabolismo , Benzaldeídos/farmacologia , Córtex Cerebral/metabolismo , Eritrócitos/efeitos dos fármacos , Fármacos Hematológicos/farmacologia , Hemoglobina Falciforme/efeitos dos fármacos , Hipóxia/metabolismo , Niacinamida/análogos & derivados , Oxigênio/metabolismo , Regulação Alostérica , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Hematócrito , Hemoglobina Falciforme/metabolismo , Camundongos , Camundongos Transgênicos , Niacinamida/farmacologia , Pressão Parcial
3.
High Alt Med Biol ; 22(3): 249-262, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34152867

RESUMO

Stewart, Glenn M., Troy J. Cross, Michael J. Joyner, Steven C. Chase, Timothy Curry, Josh Lehrer-Graiwer, Kobina Dufu, Nicholas E. Vlahakis, and Bruce D. Johnson. Impact of pharmacologically left shifting the oxygen-hemoglobin dissociation curve on arterial blood gases and pulmonary gas exchange during maximal exercise in hypoxia. High Alt Med Biol. 22:249-262, 2021. Introduction: Physiological and pathological conditions, which reduce the loading of oxygen onto hemoglobin (Hb), can impair exercise capacity and cause debilitating symptoms. Accordingly, this study examined the impact of pharmacologically left shifting the oxygen-hemoglobin dissociation curve (ODC) on arterial oxygen saturation (SaO2) and exercise capacity. Methods: Eight healthy subjects completed a maximal incremental exercise test in hypoxia (FIO2: 0.125) and normoxia (FIO2: 0.21) before (Day 1) and after (Day 15) daily ingestion of 900 mg of voxelotor (an oxygen/Hb affinity modulator). Pulmonary gas exchange and arterial blood gases were assessed throughout exercise and at peak. Data for a 1,500 mg daily drug dose are reported in a limited cohort (n = 3). Results: Fourteen days of drug administration left shifted the ODC (p50 measured under standard conditions, Day 1: 28.0 ± 2.1 mmHg vs. Day 15: 26.1 ± 1.8 mmHg, p < 0.05). Throughout incremental exercise in hypoxia, SaO2 was systematically higher after drug (peak exercise SaO2 on Day 1: 71 ± 2 vs. Day 15: 81% ± 2%, p < 0.001), whereas oxygen extraction (Ca-vO2 diff) and consumption (VO2) were similar (peak exercise Ca-vO2 diff on Day 1: 11.5 ± 1.7 vs. Day 15: 11.0 ± 1.8 ml/100 ml blood, p = 0.417; peak VO2 on Day 1: 2.59 ± 0.39 vs. Day 15: 2.47 ± 0.43 l/min, p = 0.127). Throughout incremental exercise in normoxia, SaO2 was systematically higher after drug, whereas peak VO2 was reduced (peak exercise SaO2 on Day 1: 93.9 ± 1.8 vs. Day 15: 95.8% ± 1.0%, p = 0.008; peak VO2 on Day 1: 3.62 ± 0.55 vs. Day 15: 3.26 ± 52 l/min, p = 0.001). Conclusion: Pharmacologically increasing the affinity of Hb for oxygen improved SaO2 during hypoxia without impacting exercise capacity; however, left shifting the ODC in healthy individuals appears detrimental to exercise capacity in normoxia. Left shifting the ODC to different magnitudes and under more chronic forms of hypoxia warrants further study.


Assuntos
Oxigênio , Troca Gasosa Pulmonar , Teste de Esforço , Hemoglobinas , Humanos , Hipóxia , Consumo de Oxigênio
4.
J Appl Physiol (1985) ; 128(6): 1467-1476, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32324473

RESUMO

Numerous pathophysiological conditions induce hypoxemia-related cardiopulmonary perturbations, decrements in exercise capacity, and debilitating symptoms. Accordingly, this study investigated the efficacy of an allosteric hemoglobin modulator (voxelotor) to enhance arterial oxygen saturation during low-intensity exercise in hypoxia. Eight normal healthy subjects (36 ± 7 yr; 73.8 ± 9.5 kg; 3 women) completed a submaximal cycling test (60 W) under normoxic ([Formula: see text]: 0.21; O2 partial pressure: 144 mmHg) and hypoxic ([Formula: see text]: 0.125; O2 partial pressure: 82 mmHg) conditions before (day 1) and after (day 15) 14 days of oral drug administration. While stationary on a cycle ergometer and during exercise, ratings of perceived exertion (RPE) and dyspnea, oxygen consumption (V̇o2), and cardiac output (Q) were measured noninvasively, while arterial blood pressure (MAP) and blood gases ([Formula: see text], [Formula: see text], and [Formula: see text]) were measured invasively. The 14-day drug administration left shifted the oxygen-hemoglobin dissociation curve (ODC; p50 measured at standard pH and Pco2; day 1: 28.0 ± 2.1 mmHg vs. day 15: 26.1 ± 1.8 mmHg, P < 0.05). RPE, dyspnea, V̇o2, Q, and MAP were not different between day 1 and day 15. [Formula: see text] was similar during normoxia on day 1 and day 15 while stationary but higher during exercise (day 1: 95.2 ± 0.4% vs. day 15: 96.6 ± 0.3%, P < 0.05). [Formula: see text] was higher during hypoxia on day 15 while stationary (day 1: 82.9 ± 3.4% vs. day 15: 90.9 ± 1.8%, P < 0.05) and during exercise (day 1: 73.6 ± 2.5% vs. day 15: 84.8 ± 2.7%, P < 0.01). [Formula: see text] and [Formula: see text]were systematically higher and lower, respectively, after drug (P < 0.01), while the alveolar-arterial oxygen difference was unchanged suggesting hyperventilation contributed to the rise in [Formula: see text]. Oral administration of voxelotor left shifted the ODC and stimulated a mild hyperventilation, leading to improved arterial oxygen saturation without altering V̇o2 and central hemodynamics during rest and low-intensity exercise. This effect was more pronounced during submaximal hypoxic exercise, when arterial desaturation was more evident. Additional studies are needed to determine the effects of voxelotor during maximal exercise and under chronic forms of hypoxia.NEW & NOTEWORTHY In humans, a novel allosteric hemoglobin-oxygen affinity modulator was administered to comprehensively examine the cardiopulmonary consequences of stabilizing a portion of the available hemoglobin in a high-oxygen affinity state during submaximal exercise in normoxia and hypoxia. Oral administration of voxelotor enhanced arterial oxygen saturation during submaximal exercise without altering oxygen consumption and central hemodynamics; however, the partial pressure of arterial carbon dioxide was reduced and the partial pressure of arterial oxygen was increased implying that hyperventilation also contributed to the increase in oxygen saturation. The preservation of arterial oxygen saturation and content was particularly evident during hypoxic submaximal exercise, when arterial desaturation typically occurs, but this did not influence arterial-venous oxygen difference.


Assuntos
Exercício Físico , Troca Gasosa Pulmonar , Adulto , Feminino , Hemoglobinas , Humanos , Hipóxia , Masculino , Oxigênio , Consumo de Oxigênio
5.
Physiol Rep ; 7(6): e14027, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30916477

RESUMO

Sickle cell anemia (SCA) is one of the commonest severe inherited disorders. Nevertheless, effective treatments remain inadequate and novel ones are avidly sought. A promising advance has been the design of novel compounds which react with hemoglobin S (HbS) to increase oxygen (O2 ) affinity and reduce sickling. One of these, voxelotor (GBT440), is currently in advanced clinical trials. A structural analogue, GBT1118, was investigated in the current work. As RBC dehydration is important in pathogenesis of SCA, the effect of GBT1118 on RBC cation permeability was also studied. Activities of Psickle , the Gardos channel and the KCl cotransporter (KCC) were all reduced. Gardos channel and KCC activities were also inhibited in RBCs treated with Ca2+ ionophore or the thiol reagent N-ethylmaleimide, indicative of direct effects on these two transport systems. Consistent with its action on RBC membrane transporters, GBT1118 significantly increased RBC hydration. RBC hemolysis was reduced in a nonelectrolyte lysis assay. Further to its direct effects on O2 affinity, GBT1118 was therefore found to reduce RBC shrinkage and fragility. Findings reveal important effects of GBT1118 on protecting sickle cells and suggest that this is approach may represent a useful therapy for amelioration of the clinical complications of SCA.


Assuntos
Anemia Falciforme/tratamento farmacológico , Antidrepanocíticos/farmacologia , Benzaldeídos/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Hemoglobina Falciforme/metabolismo , Hemólise/efeitos dos fármacos , Niacinamida/análogos & derivados , Oxigênio/sangue , Anemia Falciforme/sangue , Anemia Falciforme/diagnóstico , Tamanho Celular/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/sangue , Niacinamida/farmacologia , Permeabilidade , Simportadores/antagonistas & inibidores , Simportadores/sangue , Cotransportadores de K e Cl-
6.
Blood ; 133(17): 1865-1875, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-30655275

RESUMO

New treatments directly targeting polymerization of sickle hemoglobin (HbS), the proximate event in the pathophysiology of sickle cell disease (SCD), are needed to address the severe morbidity and early mortality associated with the disease. Voxelotor (GBT440) is a first-in-class oral therapy specifically developed to treat SCD by modulating the affinity of hemoglobin (Hb) for oxygen, thus inhibiting HbS polymerization and downstream adverse effects of hemolytic anemia and vaso-occlusion. GBT440-001 was a phase 1/2 randomized, double-blind, placebo-controlled, single and multiple ascending dose study of voxelotor in adult healthy volunteers and patients with SCD, followed by a single-arm, open-label extension study. This report describes results of voxelotor (500-1000 mg per day) in patients with sickle cell anemia. The study evaluated the safety, tolerability, pharmacokinetic, and pharmacodynamic properties of voxelotor and established proof of concept by improving clinical measures of anemia, hemolysis, and sickling. Thirty-eight patients with SCD received 28 days of voxelotor 500, 700, or 1000 mg per day or placebo; 16 patients received 90 days of voxelotor 700 or 900 mg per day or placebo. Four patients from the 90-day cohort were subsequently enrolled in an extension study and treated with voxelotor 900 mg per day for 6 months. All patients who received multiple doses of voxelotor for ≥28 days experienced hematologic improvements including increased Hb and reduction in hemolysis and percentage of sickled red cells, supporting the potential of voxelotor to serve as a disease-modifying therapy for SCD. Voxelotor was well tolerated with no treatment-related serious adverse events and no evidence of tissue hypoxia. These trials were registered at www.clinicaltrials.gov as #NCT02285088 and #NCT03041909.


Assuntos
Anemia Falciforme/tratamento farmacológico , Benzaldeídos/uso terapêutico , Fármacos Hematológicos/uso terapêutico , Pirazinas/uso terapêutico , Pirazóis/uso terapêutico , Adolescente , Adulto , Benzaldeídos/farmacocinética , Estudos de Casos e Controles , Estudos de Coortes , Método Duplo-Cego , Feminino , Seguimentos , Fármacos Hematológicos/farmacocinética , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Prognóstico , Pirazinas/farmacocinética , Pirazóis/farmacocinética , Distribuição Tecidual , Adulto Jovem
7.
Hematol Rep ; 10(2): 7419, 2018 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-30046411

RESUMO

Sickle cell disease is characterized by hemolytic anemia, vasoocclusion and early mortality. Polymerization of hemoglobin S followed by red blood cell sickling and subsequent vascular injury are key events in the pathogenesis of sickle cell disease. Sickled red blood cells are major contributors to the abnormal blood rheology, poor microvascular blood flow and endothelial injury in sickle cell disease. Therefore, an agent that can prevent and or reverse sickling of red blood cells, may provide therapeutic benefit for the treatment of sickle cell disease. We report here that GBT440, an anti-polymerization agent being developed for the chronic treatment of sickle cell disease, increases hemoglobin oxygen affinity and reverses in vitro sickling of previously sickled red blood cells under hypoxic conditions. Our results suggest that besides preventing sickling of red blood cells, GBT440 may mitigate vasoocclusion and microvascular dysfunction by reversing sickling of circulating sickled red blood cells in vivo.

8.
Clin Hemorheol Microcirc ; 70(1): 95-105, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29660913

RESUMO

BACKGROUND: In sickle cell disease (SCD), polymerization of hemoglobin S (HbS) leads to the formation of rigid, non-deformable sickled RBCs. Loss of RBC deformability, sickling and irreversible membrane damage causes abnormal blood rheology, and increases viscosity which contributes to vasoocclusion and other SCD pathophysiology. GBT440 (generic name voxelotor) is a novel anti-polymerization and anti-sickling agent currently undergoing clinical evaluation for the treatment of SCD. OBJECTIVE: The purpose of this study was to determine the effects of GBT440 on deformability of sickle RBCs (SS RBCs) and the hyperviscosity of sickle cell blood (SS blood). METHODS: The mechanical and rheological properties of GBT440-treated SS RBCs were measured using micropipette and filtration techniques. The viscosity of sickle blood was measured using a Wells-Brookfield cone/plate viscometer. RESULTS: GBT440 restored movement of deoxygenated SS RBCs through a gel filtration column and reduced the pressure required to pass SS RBCs through a polycarbonate filter. Moreover, GBT440 decreased the membrane shear elastic modulus of SS RBCs assessed via micropipette aspiration and reduced the hyperviscosity of SS blood under deoxygenated conditions. CONCLUSIONS: GBT440 maintains SS RBC deformability and improves SS blood viscosity by inhibiting HbS polymerization under deoxygenated conditions. These results further support development of GBT440 as a disease-modifying agent in SCD patients.


Assuntos
Anemia Falciforme/sangue , Viscosidade Sanguínea/genética , Deformação Eritrocítica/fisiologia , Eritrócitos Anormais/fisiologia , Humanos
9.
J Appl Physiol (1985) ; 124(4): 899-905, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357510

RESUMO

Acute respiratory distress syndrome (ARDS) is characterized by lung inflammation and pulmonary edema, leading to arterial hypoxemia and death if the hypoxemia is severe. Strategies to correct hypoxemia have the potential to improve clinical outcomes in ARDS. The goal of this study was to evaluate the potential of hemoglobin modification as a novel therapy for ARDS-induced hypoxemia. The therapeutic effect of two different doses of GBT1118, a compound that increases the oxygen affinity of hemoglobin, was evaluated in a murine model of acute lung injury induced by intratracheal LPS instillation 24 h before exposure to 5% or 10% hypoxia ( n = 8-15 per group). As expected, administration of GBT1118 to mice significantly increased the oxygen affinity of hemoglobin. Compared with mice receiving vehicle control, mice treated with GBT1118 had significantly lower mortality after LPS + 5% hypoxia (47% with vehicle vs. 22% with low-dose GBT1118, 13% with high-dose GBT1118, P = 0.032 by log rank) and had reduced severity of illness. Mice treated with GBT1118 showed a sustained significant increase in SpO2 over 4 h of hypoxia exposure. Treatment with GBT1118 did not alter alveolar-capillary permeability, bronchoalveolar lavage (BAL) inflammatory cell counts, or BAL concentrations of IL-1ß, TNF-α, or macrophage inflammatory protein-1α. High-dose GBT1118 did not affect histological lung injury but did decrease tissue hypoxia as measured intensity of pimonidazole (Hypoxyprobe) staining in liver ( P = 0.043) and kidney ( P = 0.043). We concluded that increasing the oxygen affinity of hemoglobin using GBT1118 may be a novel therapy for treating hypoxemia associated with acute lung injury. NEW & NOTEWORTHY In this study, we show that GBT1118, a compound that increases hemoglobin affinity for oxygen, improves survival and oxygen saturation in a two-hit lung injury model of intratracheal LPS without causing tissue hypoxia. Modulation of hemoglobin oxygen affinity represents a novel therapeutic approach to treatment of acute lung injury and acute respiratory distress syndrome, conditions characterized by hypoxemia.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Benzaldeídos/uso terapêutico , Niacinamida/análogos & derivados , Lesão Pulmonar Aguda/etiologia , Animais , Benzaldeídos/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hipóxia/complicações , Hipóxia/tratamento farmacológico , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Niacinamida/farmacologia , Niacinamida/uso terapêutico
10.
Am J Physiol Heart Circ Physiol ; 313(2): H381-H391, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526710

RESUMO

Adaptation to hypoxia requires compensatory mechanisms that affect O2 transport and utilization. Decreased hemoglobin (Hb) O2 affinity is considered part of the physiological adaptive process to chronic hypoxia. However, this study explores the hypothesis that increased Hb O2 affinity can complement acute physiological responses to hypoxia by increasing O2 uptake and delivery compared with normal Hb O2 affinity during acute severe hypoxia. To test this hypothesis, Hb O2 affinity in mice was increased by oral administration of 2-hydroxy-6-{[(2S)-1-(pyridine-3-carbonyl)piperidin-2yl] methoxy}benzaldehyde (GBT1118; 70 or 140 mg/kg). Systemic and microcirculatory hemodynamics and oxygenation parameters were studied during hypoxia in awake-instrumented mice. GBT1118 increased Hb O2 affinity and decreased the Po2 at which 50% of Hb is saturated with O2 (P50) from 43 ± 1.1 to 18.3 ± 0.9 mmHg (70 mg/kg) and 7.7 ± 0.2 mmHg (140 mg/kg). In a dose-dependent fashion, GBT1118 increased arterial O2 saturation by 16% (70 mg/kg) and 40% (140 mg/kg) relative to the control group during 5% O2 hypoxia. In addition, a GBT1118-induced increase in Hb O2 affinity reduced hypoxia-induced hypotension compared with the control group. Moreover, microvascular blood flow was higher during hypoxia in GBT1118-treated groups than the control group. The increased O2 saturation and improved blood flow in GBT1118-treated groups preserved higher interstitial tissue Po2 than in the control group during 5% O2 hypoxia. In conclusion, increased Hb O2 affinity enhanced physiological tolerance to hypoxia, as evidenced by improved hemodynamics and tissue oxygenation. Therefore, pharmacologically induced increases in Hb O2 affinity become a potential therapeutic approach to improve tissue oxygenation in pulmonary diseases characterized by severe hypoxemia.NEW & NOTEWORTHY This study establishes that pharmacological modification of hemoglobin O2 affinity can be a promising and novel therapeutic strategy for the treatment of hypoxic hypoxia and paves the way for the clinical development of molecules that prevent hypoxemia.


Assuntos
Benzaldeídos/farmacologia , Hipóxia/tratamento farmacológico , Niacinamida/análogos & derivados , Oxigênio/sangue , Oxiemoglobinas/metabolismo , Pele/irrigação sanguínea , Adaptação Fisiológica , Administração Oral , Animais , Benzaldeídos/administração & dosagem , Benzaldeídos/farmacocinética , Biomarcadores/sangue , Velocidade do Fluxo Sanguíneo , Pressão Sanguínea , Modelos Animais de Doenças , Frequência Cardíaca , Hipóxia/sangue , Hipóxia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Microcirculação/efeitos dos fármacos , Niacinamida/administração & dosagem , Niacinamida/farmacocinética , Niacinamida/farmacologia , Fluxo Sanguíneo Regional , Índice de Gravidade de Doença
11.
ACS Med Chem Lett ; 8(3): 321-326, 2017 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-28337324

RESUMO

We report the discovery of a new potent allosteric effector of sickle cell hemoglobin, GBT440 (36), that increases the affinity of hemoglobin for oxygen and consequently inhibits its polymerization when subjected to hypoxic conditions. Unlike earlier allosteric activators that bind covalently to hemoglobin in a 2:1 stoichiometry, 36 binds with a 1:1 stoichiometry. Compound 36 is orally bioavailable and partitions highly and favorably into the red blood cell with a RBC/plasma ratio of ∼150. This partitioning onto the target protein is anticipated to allow therapeutic concentrations to be achieved in the red blood cell at low plasma concentrations. GBT440 (36) is in Phase 3 clinical trials for the treatment of sickle cell disease (NCT03036813).

12.
Hematol Rep ; 8(3): 6637, 2016 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-27757216

RESUMO

In sickle cell trait (SCT), hemoglobin A (HbA) and S (HbS) are co-expressed in each red blood cell (RBC). While homozygous expression of HbS (HbSS) leads to polymerization and sickling of RBCs resulting in sickle cell disease (SCD) characterized by hemolytic anemia, painful vaso-occlusive episodes and shortened life-span, SCT is considered a benign condition usually with minor or no complications related to sickling. However, physical activities that cause increased tissue oxygen demand, dehydration and/or metabolic acidosis leads to increased HbS polymerization and life-threatening complications including death. We report that GBT440, an agent being developed for the treatment of SCD, increases the affinity of oxygen for Hb and inhibits in vitro polymerization of a mixture of HbS and HbA that simulates SCT blood. Moreover, GBT440 prevents sickling of SCT blood under in vitro conditions mimicking strenuous exercise with hypoxia, dehydration and acidosis. Together, our results indicate that GBT440 may have the potential to protect SCT individuals from sickling-related complications during conditions that favor HbS polymerization.

13.
Physiol Rep ; 4(17)2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27624688

RESUMO

Although exertional dyspnea and worsening hypoxia are hallmark clinical features of idiopathic pulmonary fibrosis (IPF), no drug currently available could treat them. GBT1118 is a novel orally bioavailable small molecule that binds to hemoglobin and produces a concentration-dependent left shift of the oxygen-hemoglobin dissociation curve with subsequent increase in hemoglobin-oxygen affinity and arterial oxygen loading. To assess whether pharmacological modification of hemoglobin-oxygen affinity could ameliorate hypoxemia associated with lung fibrosis, we evaluated GBT1118 in a bleomycin-induced mouse model of hypoxemia and fibrosis. After pulmonary fibrosis and hypoxemia were induced, GBT1118 was administered for eight consecutive days. Hypoxemia was determined by monitoring arterial oxygen saturation, while the severity of pulmonary fibrosis was assessed by histopathological evaluation and determination of collagen and leukocyte levels in bronchoalveolar lavage fluid. We found that hemoglobin modification by GBT1118 had strong antihypoxemic therapeutic effects with improved arterial oxygen saturation to near normal level. Moreover, GBT1118 treatment significantly attenuated bleomycin-induced lung fibrosis, collagen accumulation, body weight loss, and leukocyte infiltration. This study is the first to suggest the beneficial effects of hemoglobin modification in fibrotic lungs and offers a promising and novel therapeutic strategy for the treatment of hypoxemia associated with chronic fibrotic lung disorders in human, including IPF.


Assuntos
Benzaldeídos/administração & dosagem , Bleomicina/administração & dosagem , Hipóxia/induzido quimicamente , Fibrose Pulmonar Idiopática/induzido quimicamente , Niacinamida/análogos & derivados , Oxigênio/metabolismo , Oxiemoglobinas/efeitos dos fármacos , Administração Oral , Animais , Benzaldeídos/metabolismo , Benzaldeídos/farmacocinética , Benzaldeídos/farmacologia , Bleomicina/efeitos adversos , Bleomicina/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Colágeno/efeitos dos fármacos , Colágeno/metabolismo , Dispneia/diagnóstico , Dispneia/etiologia , Hipóxia/complicações , Hipóxia/tratamento farmacológico , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Niacinamida/administração & dosagem , Niacinamida/metabolismo , Niacinamida/farmacocinética , Niacinamida/farmacologia , Oxigênio/sangue , Oxiemoglobinas/metabolismo , Distribuição Aleatória
14.
Br J Haematol ; 175(1): 141-53, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27378309

RESUMO

A major driver of the pathophysiology of sickle cell disease (SCD) is polymerization of deoxygenated haemoglobin S (HbS), which leads to sickling and destruction of red blood cells (RBCs) and end-organ damage. Pharmacologically increasing the proportion of oxygenated HbS in RBCs may inhibit polymerization, prevent sickling and provide long term disease modification. We report that GBT440, a small molecule which binds to the N-terminal α chain of Hb, increases HbS affinity for oxygen, delays in vitro HbS polymerization and prevents sickling of RBCs. Moreover, in a murine model of SCD, GBT440 extends the half-life of RBCs, reduces reticulocyte counts and prevents ex vivo RBC sickling. Importantly, oral dosing of GBT440 in animals demonstrates suitability for once daily dosing in humans and a highly selective partitioning into RBCs, which is a key therapeutic safety attribute. Thus, GBT440 has the potential for clinical use as a disease-modifying agent in sickle cell patients.


Assuntos
Anemia Falciforme/metabolismo , Antidrepanocíticos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Eritrócitos Anormais/efeitos dos fármacos , Eritrócitos Anormais/metabolismo , Hemoglobina Falciforme/metabolismo , Oxigênio/metabolismo , Anemia Falciforme/sangue , Anemia Falciforme/tratamento farmacológico , Animais , Antidrepanocíticos/química , Antidrepanocíticos/farmacocinética , Gasometria , Modelos Animais de Doenças , Hemoglobina Falciforme/química , Humanos , Camundongos , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/metabolismo , Ligação Proteica
15.
PLoS One ; 7(8): e43804, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22928037

RESUMO

The conserved TREX complex, which contains UAP56, Aly, CIP29, and the multi-subunit THO complex, functions in mRNA export. Recently, several putative new components of the human TREX complex were identified by mass spectrometry. Here, we investigated the function of two of these, PDIP3 and ZC11A. Our data indicate that both of these proteins are components of a common TREX complex and function in mRNA export. Recently, we found that both CIP29 and Aly associate with the DEAD box helicase UAP56 and with the TREX complex in an ATP-dependent manner. We now show that this is also the case for PDIP3 and ZC11A. Thus, together with previous work, our data indicate that the TREX complex participates in multiple ATP-dependent interactions.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Transporte de RNA , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/química , Células HeLa , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Ligação Proteica , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Dedos de Zinco
16.
Nat Commun ; 1: 97, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20981025

RESUMO

The TREX complex, which functions in mRNA export, is recruited to mRNA during splicing. Both the splicing machinery and the TREX complex are concentrated in 20-50 discrete foci known as nuclear speckle domains. In this study, we use a model system where DNA constructs are microinjected into HeLa cell nuclei, to follow the fates of the transcripts. We show that transcripts lacking functional splice sites, which are inefficiently exported, do not associate with nuclear speckle domains but are instead distributed throughout the nucleoplasm. In contrast, pre-mRNAs containing functional splice sites accumulate in nuclear speckles, and our data suggest that splicing occurs in these domains. When the TREX components UAP56 or Aly are knocked down, spliced mRNA, as well as total polyA+ RNA, accumulates in nuclear speckle domains. Together, our data raise the possibility that pre-mRNA undergoes splicing in nuclear speckle domains, before their release by TREX components for efficient export to the cytoplasm.


Assuntos
Splicing de RNA/genética , RNA Mensageiro/genética , RNA Helicases DEAD-box/genética , Células HeLa , Humanos , Hibridização in Situ Fluorescente , Proteínas Nucleares/genética , Interferência de RNA , Precursores de RNA/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética
17.
Genes Dev ; 24(18): 2043-53, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844015

RESUMO

The conserved TREX mRNA export complex is known to contain UAP56, Aly, Tex1, and the THO complex. Here, we carried out proteomic analysis of immunopurified human TREX complex and identified the protein CIP29 as the only new component with a clear yeast relative (known as Tho1). Tho1 is known to function in mRNA export, and we provide evidence that CIP29 likewise functions in this process. Like the known TREX components, a portion of CIP29 localizes in nuclear speckle domains, and its efficient recruitment to mRNA is both splicing- and cap-dependent. We show that UAP56 mediates an ATP-dependent interaction between the THO complex and both CIP29 and Aly, indicating that TREX assembly is ATP-dependent. Using recombinant proteins expressed in Escherichia coli, we show that UAP56, Aly, and CIP29 form an ATP-dependent trimeric complex, and UAP56 bridges the interaction between CIP29 and Aly. We conclude that the interaction of two conserved export proteins, CIP29 and Aly, with UAP56 is strictly regulated by ATP during assembly of the TREX complex.


Assuntos
Trifosfato de Adenosina/metabolismo , RNA Helicases DEAD-box/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células COS , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , RNA Helicases DEAD-box/genética , Exodesoxirribonucleases/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/genética , Processamento Pós-Transcricional do RNA/fisiologia , Transporte de RNA
18.
Cell ; 127(7): 1389-400, 2006 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-17190602

RESUMO

Pre-mRNAs undergo splicing to remove introns, and the spliced mRNA is exported to the cytoplasm for translation. Here we investigated the mechanism for recruitment of the conserved mRNA export machinery (TREX complex) to mRNA. We show that the human TREX complex is recruited to a region near the 5' end of mRNA, with the TREX component Aly bound closest to the 5' cap. Both TREX recruitment and mRNA export require the cap, and these roles for the cap are splicing dependent. CBP80, which is bound to the cap, associates efficiently with TREX, and Aly mediates this interaction. Together, these data indicate that the CBP80-Aly interaction results in recruitment of TREX to the 5' end of mRNA, where it functions in mRNA export. As a consequence, the mRNA would be exported in a 5' to 3' direction through the nuclear pore, as observed in early electron micrographs of giant Balbiani ring mRNPs.


Assuntos
Splicing de RNA , Transporte de RNA , RNA Mensageiro/genética , Éxons , Humanos , Modelos Genéticos , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo , Proteínas Nucleares/metabolismo , RNA Mensageiro/química , Proteínas de Ligação a RNA/metabolismo , Spliceossomos/metabolismo , Fatores de Transcrição/metabolismo
19.
Genes Dev ; 20(9): 1100-9, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651655

RESUMO

The pathway of gene expression in higher eukaryotes involves a highly complex network of physical and functional interactions among the different machines involved in each step of the pathway. Here we established an efficient in vitro system to determine how RNA polymerase II (RNAP II) transcription is functionally coupled to pre-mRNA splicing. Strikingly, our data show that nascent pre-messenger RNA (pre-mRNA) synthesized by RNAP II is immediately and quantitatively directed into the spliceosome assembly pathway. In contrast, nascent pre-mRNA synthesized by T7 RNA polymerase is quantitatively assembled into the nonspecific H complex, which consists of heterogeneous nuclear ribonucleoprotein (hnRNP) proteins and is inhibitory for spliceosome assembly. Consequently, RNAP II transcription results in a dramatic increase in both the kinetics of splicing and overall yield of spliced mRNA relative to that observed for T7 transcription. We conclude that RNAP II mediates the functional coupling of transcription to splicing by directing the nascent pre-mRNA into spliceosome assembly, thereby bypassing interaction of the pre-mRNA with the inhibitory hnRNP proteins.


Assuntos
RNA Polimerase II/metabolismo , Precursores de RNA/metabolismo , Splicing de RNA , Spliceossomos/fisiologia , Transcrição Gênica , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Células HeLa , Humanos , RNA Polimerase II/genética , Precursores de RNA/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...