Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.194
Filtrer
1.
Science ; 385(6704): 91-99, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38963839

RÉSUMÉ

Sickle cell disease (SCD) is a prevalent, life-threatening condition attributable to a heritable mutation in ß-hemoglobin. Therapeutic induction of fetal hemoglobin (HbF) can ameliorate disease complications and has been intently pursued. However, safe and effective small-molecule inducers of HbF remain elusive. We report the discovery of dWIZ-1 and dWIZ-2, molecular glue degraders of the WIZ transcription factor that robustly induce HbF in erythroblasts. Phenotypic screening of a cereblon (CRBN)-biased chemical library revealed WIZ as a previously unknown repressor of HbF. WIZ degradation is mediated by recruitment of WIZ(ZF7) to CRBN by dWIZ-1, as resolved by crystallography of the ternary complex. Pharmacological degradation of WIZ was well tolerated and induced HbF in humanized mice and cynomolgus monkeys. These findings establish WIZ degradation as a globally accessible therapeutic strategy for SCD.


Sujet(s)
Drépanocytose , Hémoglobine foetale , Hémoglobine foetale/génétique , Hémoglobine foetale/métabolisme , Animaux , Drépanocytose/génétique , Drépanocytose/métabolisme , Humains , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéolyse , Macaca fascicularis , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Cristallographie aux rayons X
2.
Biomed Pharmacother ; 176: 116849, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823275

RÉSUMÉ

Sickle cell disease (SCD) is the most severe monogenic hemoglobinopathy caused by a single genetic mutation that leads to repeated polymerization and depolymerization of hemoglobin resulting in intravascular hemolysis, cell adhesion, vascular occlusion, and ischemia-reperfusion injury. Hemolysis causes oxidative damage indirectly by generating reactive oxygen species through various pathophysiological mechanisms, which include hemoglobin autoxidation, endothelial nitric oxide synthase uncoupling, reduced nitric oxide bioavailability, and elevated levels of asymmetric dimethylarginine. Red blood cells have a built-in anti-oxidant system that includes enzymes like sodium dismutase, catalase, and glutathione peroxidase, along with free radical scavenging molecules, such as vitamin C, vitamin E, and glutathione, which help them to fight oxidative damage. However, these anti-oxidants may not be sufficient to prevent the effects of oxidative stress in SCD patients. Therefore, in line with a recent FDA request that the focus to be placed on the development of innovative therapies for SCD that address the root cause of the disease, there is a need for therapies that target oxidative stress and restore redox balance in SCD patients. This review summarizes the current state of knowledge regarding the role of oxidative stress in SCD and the potential benefits of anti-oxidant therapies. It also discusses the challenges and limitations of these therapies and suggests future directions for research and development.


Sujet(s)
Drépanocytose , Antioxydants , Stress oxydatif , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Antioxydants/usage thérapeutique , Antioxydants/pharmacologie , Animaux , Espèces réactives de l'oxygène/métabolisme
3.
Blood Adv ; 8(12): 3272-3283, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38640339

RÉSUMÉ

ABSTRACT: Sickle cell disease (SCD) is a hereditary hemoglobinopathy marked by hemolytic anemia and vaso-occlusive events (VOEs). Chronic endothelial activation, inflammation, and coagulation activation contribute to vascular congestion, VOEs, and end-organ damage. Coagulation proteases such as thrombin and activated protein C (APC) modulate inflammation and endothelial dysfunction by activating protease-activated receptor 1 (PAR1), a G-protein-coupled receptor. Thrombin cleaves PAR1 at Arg41, while APC cleaves PAR1 at Arg46, initiating either proinflammatory or cytoprotective signaling, respectively, a signaling conundrum known as biased agonism. Our prior research established the role of thrombin and PAR1 in vascular stasis in an SCD mouse model. However, the role of APC and APC-biased PAR1 signaling in thrombin generation, inflammation, and endothelial activation in SCD remains unexplored. Inhibition of APC in SCD mice increased thrombin generation, inflammation, and endothelial activation during both steady state and tumor necrosis factor α challenge. To dissect the individual contributions of thrombin-PAR1 and APC-PAR1 signaling, we used transgenic mice with point mutations at 2 PAR1 cleavage sites, ArgR41Gln (R41Q) imparting insensitivity to thrombin and Arg46Gln (R46Q) imparting insensitivity to APC. Sickle bone marrow chimeras expressing PAR1-R41Q exhibited reduced thrombo-inflammatory responses compared with wild type PAR1 or PAR1-R46Q mice. These findings highlight the potential benefit of reducing thrombin-dependent PAR1 activation while preserving APC-PAR1 signaling in SCD thromboinflammation. These results also suggest that pharmacological strategies promoting biased PAR1 signaling could effectively mitigate vascular complications associated with SCD.


Sujet(s)
Drépanocytose , Modèles animaux de maladie humaine , Inflammation , Protéine C , Récepteur de type PAR-1 , Thrombine , Animaux , Drépanocytose/métabolisme , Drépanocytose/complications , Récepteur de type PAR-1/métabolisme , Souris , Protéine C/métabolisme , Inflammation/métabolisme , Thrombine/métabolisme , Transduction du signal , Souris transgéniques , Thrombose/métabolisme , Thrombose/étiologie , Humains
4.
Toxicol In Vitro ; 98: 105832, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38653437

RÉSUMÉ

Sickle cell disease (SCD) is a hereditary hemoglobinopathy, caused by a mutation at position 6 of the ß-globin chain and patients are frequently exposed to several blood transfusions in order to maintain physiological function. Transfusion blood bags are composed of PVC and phthalates (as DEHP) are often introduced to the material in order to confer malleability. In this sense, DEHP can easily elute to the blood and cause harmful effects. This study aimed to unravel DEHP effect on SCD patient's hemoglobin function. We found that HbS polymerization using whole erythrocytes is decreased by DEHP in ex vivo experiments and this effect might be mediated by the DEHP-VAL6 interaction, evaluated in silico. Isolated HbS exhibited less polymerization at low DEHP concentrations and increased polymerization rate at higher concentration. When analyzing the propensity to aggregate, HbS is more inclined to aggregate when compared to HbA due to the residue 6 mutation. Circular dichroism showed characteristic hemoglobin peaks for oxygenated HbS that are lost when oxygen is sequestered, and DEHP at higher concentration mildly recovers a peak close to the second hemoglobin one. Finally, by transmission electron microscopy we demonstrated that high DEHP concentration increased polymer formation with a more organized structure. These findings show for the first-time the beneficial effect of low-dose DEHP on HbS polymerization.


Sujet(s)
Drépanocytose , Phtalate de bis[2-éthylhexyle] , Érythrocytes , Hémoglobine S , Polymérisation , Humains , Drépanocytose/génétique , Drépanocytose/métabolisme , Hémoglobine S/génétique , Hémoglobine S/métabolisme , Érythrocytes/effets des médicaments et des substances chimiques , Érythrocytes/métabolisme , Phtalate de bis[2-éthylhexyle]/toxicité , Simulation numérique
5.
Blood Cells Mol Dis ; 105: 102824, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38262104

RÉSUMÉ

In preparation for hematopoietic stem cell mobilization and collection, current ex vivo gene therapy protocols for sickle cell disease require patients to undergo several months of chronic red cell transfusion. For health care equity, alternatives to red cell transfusion should be available. We examined whether treatment with GBT1118, the murine analog of voxelotor, could be a safe and feasible alternative to red cell transfusion. We found that 3 weeks of treatment with GBT1118 increased the percentage of bone marrow hematopoietic stem cells and upon plerixafor mobilization, the percentage of peripheral blood hematopoietic stem cells. Our data suggest that voxelotor should be further explored for its potential safety and utility as preparation for hematopoietic stem cell mobilization and collection.


Sujet(s)
Drépanocytose , Benzaldéhydes , Transplantation de cellules souches hématopoïétiques , Composés hétérocycliques , Nicotinamide/analogues et dérivés , Pyrazines , Humains , Souris , Animaux , Mobilisation de cellules souches hématopoïétiques/méthodes , Moelle osseuse/métabolisme , Cellules souches hématopoïétiques/métabolisme , Composés hétérocycliques/usage thérapeutique , Composés hétérocycliques/pharmacologie , Pyrazoles , Drépanocytose/génétique , Drépanocytose/thérapie , Drépanocytose/métabolisme , Thérapie génétique/effets indésirables , Facteur de stimulation des colonies de granulocytes/pharmacologie
6.
Exp Hematol ; 131: 104153, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38237718

RÉSUMÉ

The formation of new red blood cells (RBC) (erythropoiesis) has served as a paradigm for understanding cellular differentiation and developmental control of gene expression. The metabolic regulation of this complex, coordinated process remains poorly understood. Each step of erythropoiesis, including lineage specification of hematopoietic stem cells, proliferation, differentiation, and terminal maturation into highly specialized oxygen-carrying cells, has unique metabolic requirements. Developing erythrocytes in mammals are also characterized by unique metabolic events such as loss of mitochondria with switch to glycolysis, ejection of nucleus and organelles, high-level heme and hemoglobin synthesis, and antioxidant requirement to protect hemoglobin molecules. Genetic defects in metabolic enzymes, including pyruvate kinase and glucose-6-phosphate dehydrogenase, cause common erythrocyte disorders, whereas other inherited disorders such as sickle cell disease and ß-thalassemia display metabolic abnormalities associated with disease pathophysiology. Here we describe recent discoveries on the metabolic control of RBC formation and function, highlight emerging concepts in understanding the erythroid metabolome, and discuss potential therapeutic benefits of targeting metabolism for RBC disorders.


Sujet(s)
Drépanocytose , Érythropoïèse , Animaux , Humains , Érythropoïèse/physiologie , Érythrocytes/métabolisme , Drépanocytose/métabolisme , Mitochondries/métabolisme , Hémoglobines , Mammifères
7.
Chem Biol Drug Des ; 103(1): e14371, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37798397

RÉSUMÉ

Sickle cell disease (SCD) is the most common genetic disorder, affecting millions of people worldwide. Aromatic aldehydes, which increase the oxygen affinity of human hemoglobin to prevent polymerization of sickle hemoglobin and inhibit red blood cell (RBC) sickling, have been the subject of keen interest for the development of effective treatment against SCD. However, the aldehyde functional group metabolic instability has severly hampered their development, except for voxelotor, which was approved in 2019 for SCD treatment. To improve the metabolic stability of aromatic aldehydes, we designed and synthesized novel molecules by incorporating Michael acceptor reactive centers into the previously clinically studied aromatic aldehyde, 5-hydroxymethylfurfural (5-HMF). Eight such derivatives, referred to as MMA compounds were synthesized and studied for their functional and biological activities. Unlike 5-HMF, which forms Schiff-base interaction with αVal1 nitrogen of hemoglobin, the MMA compounds covalently interacted with ßCys93, as evidenced by reverse-phase HPLC and disulfide exchange reaction, explaining their RBC sickling inhibitory activities, which at 2 mM and 5 mM, range from 0% to 21% and 9% to 64%, respectively. Additionally, the MMA compounds showed a second mechanism of sickling inhibition (12%-41% and 13%-62% at 2 mM and 5 mM, respectively) by directly destabilizing the sickle hemoglobin polymer. In vitro studies demonstrated sustained pharmacologic activities of the compounds compared to 5-HMF. These findings hold promise for advancing SCD therapeutics.


Sujet(s)
Drépanocytose , Antidrépanocytaires , Humains , Antidrépanocytaires/pharmacologie , Antidrépanocytaires/usage thérapeutique , Hémoglobines/métabolisme , Hémoglobines/usage thérapeutique , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Hémoglobine S/métabolisme , Hémoglobine S/usage thérapeutique , Furanes , Aldéhydes/usage thérapeutique , Oxygène/métabolisme
8.
Blood ; 143(6): 535-547, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-37992231

RÉSUMÉ

ABSTRACT: In humans, ∼0.1% to 0.3% of circulating red blood cells (RBCs) are present as platelet-RBC (P-RBC) complexes, and it is 1% to 2% in mice. Excessive P-RBC complexes are found in diseases that compromise RBC health (eg, sickle cell disease and malaria) and contribute to pathogenesis. However, the physiological role of P-RBC complexes in healthy blood is unknown. As a result of damage accumulated over their lifetime, RBCs nearing senescence exhibit physiological and molecular changes akin to those in platelet-binding RBCs in sickle cell disease and malaria. Therefore, we hypothesized that RBCs nearing senescence are targets for platelet binding and P-RBC formation. Confirming this hypothesis, pulse-chase labeling studies in mice revealed an approximately tenfold increase in P-RBC complexes in the most chronologically aged RBC population compared with younger cells. When reintroduced into mice, these complexes were selectively cleared from the bloodstream (in preference to platelet-free RBC) through the reticuloendothelial system and erythrophagocytes in the spleen. As a corollary, patients without a spleen had higher levels of complexes in their bloodstream. When the platelet supply was artificially reduced in mice, fewer RBC complexes were formed, fewer erythrophagocytes were generated, and more senescent RBCs remained in circulation. Similar imbalances in complex levels and senescent RBC burden were observed in humans with immune thrombocytopenia (ITP). These findings indicate that platelets are important for binding and clearing senescent RBCs, and disruptions in platelet count or complex formation and clearance may negatively affect RBC homeostasis and may contribute to the known risk of thrombosis in ITP and after splenectomy.


Sujet(s)
Drépanocytose , Paludisme , Thrombopénie , Humains , Animaux , Souris , Sujet âgé , Plaquettes/métabolisme , Érythrocytes/métabolisme , Thrombopénie/métabolisme , Drépanocytose/métabolisme
9.
Rapid Commun Mass Spectrom ; 38(2): e9671, 2024 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-38124165

RÉSUMÉ

RATIONALE: Sickle cell disease, a debilitating genetic disorder affecting numerous newborns globally, has historically received limited attention in pharmaceutical research. However, recent years have witnessed a notable shift, with the Food and Drug Administration approving three innovative disease-modifying medications. Voxelotor, also known as GBT440, is a promising compound that effectively prevents sickling, providing a safe approach to alleviate chronic hemolytic anemia in sickle cell disease. It is a novel, orally bioavailable small molecule that inhibits hemoglobin S polymerization by enhancing oxygen affinity to hemoglobin. The investigation demonstrated that voxelotor led to an unintended elevation of hemoglobin levels in healthy individuals by increasing serum erythropoietin levels. METHODS: Voxelotor and its metabolites in an in vitro setting utilizing equine liver microsomes were discussed. Plausible structures of the identified metabolites were inferred through the application of liquid chromatography in conjunction with high-resolution mass spectrometry. RESULTS: Under the experimental conditions, a total of 31 metabolites were detected, including 16 phase I metabolites, two phase II metabolites, and 13 conjugates of phase I metabolites. The principal phase I metabolites were generated through processes such as hydroxylation, reduction, and dissociation. The presence of glucuronide and sulfate conjugates of the parent drug were also observed, along with hydroxylated, reduced, and dissociated analogs. CONCLUSIONS: The data acquired will accelerate the identification of voxelotor and related compounds, aiding in the detection of their illicit use in competitive sports. It is crucial to emphasize that the metabolites detailed in this manuscript were identified through in vitro experiments and their detection in an in vivo study may not be guaranteed.


Sujet(s)
Drépanocytose , Dopage sportif , Nouveau-né , Humains , Animaux , Equus caballus , Hémoglobine S/composition chimique , Hémoglobine S/métabolisme , Hémoglobine S/usage thérapeutique , Dopage sportif/prévention et contrôle , Polymérisation , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Benzaldéhydes/pharmacologie , Benzaldéhydes/usage thérapeutique , Hémoglobines
10.
Blood Cells Mol Dis ; 104: 102792, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37633023

RÉSUMÉ

Sickle cell disease (SCD) is the most common ß-hemoglobinopathy caused by various mutations in the adult ß-globin gene resulting in sickle hemoglobin production, chronic hemolytic anemia, pain, and progressive organ damage. The best therapeutic strategies to manage the clinical symptoms of SCD is the induction of fetal hemoglobin (HbF) using chemical agents. At present, among the Food and Drug Administration-approved drugs to treat SCD, hydroxyurea is the only one proven to induce HbF protein synthesis, however, it is not effective in all people. Therefore, we evaluated the ability of the novel Bach1 inhibitor, HPP-D to induce HbF in KU812 cells and primary sickle erythroid progenitors. HPP-D increased HbF and decreased Bach1 protein levels in both cell types. Furthermore, chromatin immunoprecipitation assay showed reduced Bach1 and increased NRF2 binding to the γ-globin promoter antioxidant response elements. We also observed increased levels of the active histone marks H3K4Me1 and H3K4Me3 supporting an open chromatin configuration. In primary sickle erythroid progenitors, HPP-D increased γ-globin transcription and HbF positive cells and reduced sickled erythroid progenitors under hypoxia conditions. Collectively, our data demonstrate that HPP-D induces γ-globin gene transcription through Bach1 inhibition and enhanced NRF2 binding in the γ-globin promoter antioxidant response elements.


Sujet(s)
Drépanocytose , Globines gamma , Humains , Drépanocytose/traitement médicamenteux , Drépanocytose/génétique , Drépanocytose/métabolisme , Hémoglobine foetale/génétique , Hémoglobine foetale/métabolisme , Globines gamma/génétique , Hémoglobine S/génétique , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/usage thérapeutique , Activation de la transcription/effets des médicaments et des substances chimiques , Cellules érythroïdes/effets des médicaments et des substances chimiques , Cellules érythroïdes/métabolisme
11.
Haematologica ; 109(6): 1918-1932, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38105727

RÉSUMÉ

Inflammatory vasculopathy is critical in sickle cell disease (SCD)-associated organ damage. An imbalance between pro-inflammatory and pro-resolving mechanisms in response to different triggers such as hypoxia/reoxygenation or infections has been proposed to contribute to the progression of SCD. Administration of specialized pro-resolving lipid mediators may provide an effective therapeutic strategy to target inflammatory vasculopathy and to modulate inflammatory response. Epeleuton (15 hydroxy eicosapentaenoic acid ethyl ester) is a novel, orally administered, second-generation ω-3 fatty acid with a favorable clinical safety profile. In this study we show that epeleuton re-programs the lipidomic pattern of target organs for SCD towards a pro-resolving pattern. This protects against systemic and local inflammatory responses and improves red cell features, resulting in reduced hemolysis and sickling compared with that in vehicle-treated SCD mice. In addition, epeleuton prevents hypoxia/reoxygenation-induced activation of nuclear factor-κB with downregulation of the NLRP3 inflammasome in lung, kidney, and liver. This was associated with downregulation of markers of vascular activation in epeleuton-treated SCD mice when compared to vehicle-treated animals. Collectively our data support the potential therapeutic utility of epeleuton and provide the rationale for the design of clinical trials to evaluate the efficacy of epeleuton in patients with SCD.


Sujet(s)
Drépanocytose , Modèles animaux de maladie humaine , Lésion d'ischémie-reperfusion , Animaux , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Drépanocytose/anatomopathologie , Drépanocytose/complications , Souris , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Acide eicosapentanoïque/analogues et dérivés , Acide eicosapentanoïque/pharmacologie , Acides gras omega-3/pharmacologie , Humains , Mâle , Hypoxie/métabolisme , Hypoxie/traitement médicamenteux
12.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-38069257

RÉSUMÉ

Hemolytic disorders, like malaria and sickle cell disease (SCD), are responsible for significant mortality and morbidity rates globally, specifically in the Americas and Africa. In both malaria and SCD, red blood cell hemolysis leads to the release of a cytotoxic heme that triggers the expression of unique inflammatory profiles, which mediate the tissue damage and pathogenesis of both diseases. MicroRNAs (miRNAs), such as miR-451a and let-7i-5p, contribute to a reduction in the pro-inflammatory responses induced by circulating free hemes. MiR-451a targets both IL-6R (pro-inflammatory) and 14-3-3ζ (anti-inflammatory), and when this miRNA is present, IL-6R is reduced and 14-3-3ζ is increased. Let-7i-5p targets and reduces TLR4, which results in anti-inflammatory signaling. These gene targets regulate inflammation via NFκB regulation and increase anti-inflammatory signaling. Additionally, they indirectly regulate the expression of key heme scavengers, such as heme-oxygenase 1 (HO-1) (coded by the HMOX1 gene) and hemopexin, to decrease circulating cytotoxic heme concentration. MiRNAs can be transported within extracellular vesicles (EVs), such as exosomes, offering insights into the mechanisms of mitigating heme-induced inflammation. We tested the hypothesis that miR-451a- or let-7i-5p-loaded artificial EVs (liposomes) will reduce heme-induced inflammation in brain vascular endothelial cells (HBEC-5i, ATCC: CRL-3245) and macrophages (THP-1, ATCC: TIB-202) in vitro. We completed arginase and nitric oxide assays to determine anti- and pro-inflammatory macrophage presence, respectively. We also assessed the gene expression of IL-6R, TLR4, 14-3-3ζ, and NFκB by RT-qPCR for both cell lines. Our findings revealed that the exposure of HBEC-5i and THP-1 to liposomes loaded with miR-451a or let-7i-5p led to a reduced mRNA expression of IL-6R, TLR4, 14-3-3ζ, and NFκB when treated with a heme. It also resulted in the increased expression of HMOX1 and hemopexin. Finally, macrophages exhibited a tendency toward adopting an anti-inflammatory differentiation phenotype. These findings suggest that miRNA-loaded liposomes can modulate heme-induced inflammation and can be used to target specific cellular pathways, mediating inflammation common to hematological conditions, like malaria and SCD.


Sujet(s)
Drépanocytose , Paludisme , microARN , Humains , microARN/métabolisme , Hémolyse , Liposomes/métabolisme , Hème/métabolisme , Cellules endothéliales/métabolisme , Hémopexine/métabolisme , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Protéines 14-3-3/métabolisme , Drépanocytose/génétique , Drépanocytose/métabolisme , Inflammation/génétique , Inflammation/métabolisme , Anti-inflammatoires/métabolisme , Paludisme/métabolisme
13.
Presse Med ; 52(4): 104202, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37944640

RÉSUMÉ

Sickle cell disease (SCD) is an hereditary disorder characterized by the production of an abnormal hemoglobin called hemoglobin S (HbS). HbS may polymerize in deoxygenated conditions, which leads to red blood cell (RBC) sickling. Sickled RBCs are more rigid and fragile, and prone to lysis. SCD patients exhibit various acute and/or chronic complications, which may affect several organs. The clinical presentation of SCD is highly variable from one patient to another and cannot be only explained by RBC sickling. Increased blood viscosity, caused by the presence of RBCs with abnormal deformability and aggregation, may increase vascular resistance and increase the risk of acute and chronic vascular complications. Chronic hemolysis results in decreased nitric oxide (NO) bioavailability which may compromise vasodilation and participate to the development of chronic vasculopathy. Furthermore, chronic hemolysis is responsible for increased inflammation and oxidative stress, which affect the vascular system and may promote the adhesion of circulating cells to endothelial cells. Extracellular vesicles and especially RBC microparticles (massively released in the context of SCD) are also at the origin of vascular damages and increased white blood cells adhesion to the endothelium, which may trigger vaso-occlusive crisis and other vascular-related complications. This review highlights the fact that SCD should not only be considered as a hematological disorder but also as a vascular disease.


Sujet(s)
Drépanocytose , Maladies vasculaires , Humains , Hémolyse , Cellules endothéliales/métabolisme , Drépanocytose/complications , Drépanocytose/métabolisme , Hémoglobine S/métabolisme , Maladies vasculaires/complications
14.
PLoS One ; 18(10): e0292706, 2023.
Article de Anglais | MEDLINE | ID: mdl-37812620

RÉSUMÉ

Sickle cell disease (SCD) is a genetic disorder that has been associated with priapism. The role of hydroxyurea, a common SCD therapy, in influencing the nitric oxide (NO)-cGMP pathway and its effect on priapism is unclear. To investigate the effect of hydroxyurea treatment on smooth muscle relaxation of corpus cavernosum induced by stimulation of the NO-cGMP pathway in SCD transgenic mice and endothelial NO synthase gene-deficient (eNOS-/-) mice, which are used as model of priapism associated with the low bioavailability of endothelial NO. Four-month-old wild-type (WT, C57BL/6), SCD transgenic, and eNOS-/- male mice were treated with hydroxyurea (100 mg/Kg/day) or its vehicle (saline) daily for three weeks via intraperitoneal injections. Concentration-response curves for acetylcholine (ACh), sodium nitroprusside (SNP), and electrical field stimulation (EFS) were generated using strips of mice corpus cavernosum. The SCD mice demonstrated an amplified CC relaxation response triggered by ACh, EFS, and SNP. The corpus cavernosum relaxation responses to SNP and EFS were found to be heightened in the eNOS-/- group. However, the hydroxyurea treatment did not alter these escalated relaxation responses to ACh, EFS, and SNP in the corpus cavernosum of the SCD group, nor the relaxation responses to EFS and SNP in the eNOS-/- group. In conclusion, hydroxyurea is not effective in treating priapism associated with SCD. It is likely that excess plasma hemoglobin and reactive oxygen species, which are reported in SCD, are reacting with NO before it binds to GCs in the smooth muscle of the corpus cavernosum, thus preventing the restoration of baseline NO/cGMP levels. Furthermore, the downregulation of eNOS in the penis may impair the pharmacological action of hydroxyurea at the endothelial level in SCD mice. This study emphasize the urgency for exploring alternative therapeutic avenues for priapism in SCD that are not hindered by high plasma hemoglobin and ROS levels.


Sujet(s)
Drépanocytose , Priapisme , Humains , Souris , Mâle , Animaux , Priapisme/étiologie , Priapisme/complications , Monoxyde d'azote/métabolisme , Hydroxy-urée/pharmacologie , Hydroxy-urée/usage thérapeutique , Souris de lignée C57BL , Pénis , Nitroprussiate/pharmacologie , Nitroprussiate/métabolisme , Drépanocytose/complications , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Souris transgéniques , Relâchement musculaire , Acétylcholine/métabolisme , Phénotype , Hémoglobines/métabolisme
15.
Blood ; 142(22): 1918-1927, 2023 11 30.
Article de Anglais | MEDLINE | ID: mdl-37774369

RÉSUMÉ

Vaso-occlusive pain episodes (VOE) cause severe pain in patients with sickle cell disease (SCD). Vaso-occlusive events promote ischemia/reperfusion pathobiology that activates complement. We hypothesized that complement activation is linked to VOE. We used cold to induce VOE in the Townes sickle homozygous for hemoglobin S (HbSS) mouse model and complement inhibitors to determine whether anaphylatoxin C5a mediates VOE. We used a dorsal skinfold chamber to measure microvascular stasis (vaso-occlusion) and von Frey filaments applied to the plantar surface of the hind paw to assess mechanical hyperalgesia in HbSS and control Townes mice homozygous for hemoglobin A (HbAA) mice after cold exposure at 10°C/50°F for 1 hour. Cold exposure induced more vaso-occlusion in nonhyperalgesic HbSS mice (33%) than in HbAA mice (11%) or HbSS mice left at room temperature (1%). Cold exposure also produced mechanical hyperalgesia as measured by paw withdrawal threshold in HbSS mice compared with that in HbAA mice or HbSS mice left at room temperature. Vaso-occlusion and hyperalgesia were associated with an increase in complement activation fragments Bb and C5a in plasma of HbSS mice after cold exposure. This was accompanied by an increase in proinflammatory NF-κB activation and VCAM-1 and ICAM-1 expression in the liver. Pretreatment of nonhyperalgesic HbSS mice before cold exposure with anti-C5 or anti-C5aR monoclonal antibodies (mAbs) decreased vaso-occlusion, mechanical hyperalgesia, complement activation, and liver inflammatory markers compared with pretreatment with control mAb. Anti-C5 or -C5aR mAb infusion also abrogated mechanical hyperalgesia in HbSS mice with ongoing hyperalgesia at baseline. These findings suggest that C5a promotes vaso-occlusion, pain, and inflammation during VOE and may play a role in chronic pain.


Sujet(s)
Drépanocytose , Trait drépanocytaire , Souris , Humains , Animaux , Hyperalgésie/étiologie , Hyperalgésie/métabolisme , Souris transgéniques , Douleur , Drépanocytose/complications , Drépanocytose/génétique , Drépanocytose/métabolisme , Trait drépanocytaire/complications , Activation du complément
16.
Int J Mol Sci ; 24(11)2023 May 31.
Article de Anglais | MEDLINE | ID: mdl-37298481

RÉSUMÉ

Beta-hemoglobinopathies are the most common genetic disorders worldwide, caused by a wide spectrum of mutations in the ß-globin locus, and associated with morbidity and early mortality in case of patient non-adherence to supportive treatment. Allogeneic transplantation of hematopoietic stem cells (allo-HSCT) used to be the only curative option, although the indispensable need for an HLA-matched donor markedly restricted its universal application. The evolution of gene therapy approaches made possible the ex vivo delivery of a therapeutic ß- or γ- globin gene into patient-derived hematopoietic stem cells followed by the transplantation of corrected cells into myeloablated patients, having led to high rates of transfusion independence (thalassemia) or complete resolution of painful crises (sickle cell disease-SCD). Hereditary persistence of fetal hemoglobin (HPFH), a syndrome characterized by increased γ-globin levels, when co-inherited with ß-thalassemia or SCD, converts hemoglobinopathies to a benign condition with mild clinical phenotype. The rapid development of precise genome editing tools (ZFN, TALENs, CRISPR/Cas9) over the last decade has allowed the targeted introduction of mutations, resulting in disease-modifying outcomes. In this context, genome editing tools have successfully been used for the introduction of HPFH-like mutations both in HBG1/HBG2 promoters or/and in the erythroid enhancer of BCL11A to increase HbF expression as an alternative curative approach for ß-hemoglobinopathies. The current investigation of new HbF modulators, such as ZBTB7A, KLF-1, SOX6, and ZNF410, further expands the range of possible genome editing targets. Importantly, genome editing approaches have recently reached clinical translation in trials investigating HbF reactivation in both SCD and thalassemic patients. Showing promising outcomes, these approaches are yet to be confirmed in long-term follow-up studies.


Sujet(s)
Drépanocytose , Hémoglobinopathies , bêta-Thalassémie , Humains , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Protéines de liaison à l'ADN/métabolisme , Hémoglobinopathies/génétique , Hémoglobinopathies/thérapie , Édition de gène/méthodes , Drépanocytose/génétique , Drépanocytose/thérapie , Drépanocytose/métabolisme , Globines gamma/génétique , Globines gamma/métabolisme , bêta-Thalassémie/génétique , bêta-Thalassémie/thérapie , bêta-Thalassémie/métabolisme
18.
Blood ; 142(4): 382-396, 2023 07 27.
Article de Anglais | MEDLINE | ID: mdl-37267508

RÉSUMÉ

Sickle cell disease (SCD) is a chronic hemolytic and systemic hypoxia condition with constant oxidative stress and significant metabolic alterations. However, little is known about the correlation between metabolic alterations and the pathophysiological symptoms. Here, we report that Nrf2, a master regulator of cellular antioxidant responses, regulates the production of the metabolite l-2-hydroxyglutarate (L2HG) to mediate epigenetic histone hypermethylation for gene expression involved in metabolic, oxidative, and ferroptotic stress responses in SCD. Mechanistically, Nrf2 was found to regulate the expression of L2HG dehydrogenase (L2hgdh) to mediate L2HG production under hypoxia. Gene expression profile analysis indicated that reactive oxygen species (ROS) and ferroptosis responses were the most significantly affected signaling pathways after Nrf2 ablation in SCD. Nrf2 silencing and L2HG supplementation sensitize human sickle erythroid cells to ROS and ferroptosis stress. The absence of Nrf2 and accumulation of L2HG significantly affect histone methylation for chromatin structure modification and reduce the assembly of transcription complexes on downstream target genes to regulate ROS and ferroptosis responses. Furthermore, pharmacological activation of Nrf2 was found to have protective effects against ROS and ferroptosis stress in SCD mice. Our data suggest a novel mechanism by which Nrf2 regulates L2HG levels to mediate SCD severity through ROS and ferroptosis stress responses, suggesting that targeting Nrf2 is a viable therapeutic strategy for ameliorating SCD symptoms.


Sujet(s)
Drépanocytose , Chromatine , Épigenèse génétique , Ferroptose , Glutarates , Facteur-2 apparenté à NF-E2 , Ferroptose/génétique , Glutarates/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Drépanocytose/génétique , Drépanocytose/métabolisme , Drépanocytose/anatomopathologie , Chromatine/métabolisme , Méthylation , Alcohol oxidoreductases/métabolisme , Animaux , Souris , Espèces réactives de l'oxygène/métabolisme , Transcription génétique , Analyse de profil d'expression de gènes
19.
Cells ; 12(8)2023 04 10.
Article de Anglais | MEDLINE | ID: mdl-37190030

RÉSUMÉ

BACKGROUND: Sickle cell disease (SCD) is a highly prevalent genetic disease caused by a point mutation in the HBB gene, which can lead to chronic hemolytic anemia and vaso-occlusive events. Patient-derived induced pluripotent stem cells (iPSCs) hold promise for the development of novel predictive methods for screening drugs with anti-sickling activity. In this study, we evaluated and compared the efficiency of 2D and 3D erythroid differentiation protocols using a healthy control and SCD-iPSCs. METHODS: iPSCs were subjected to hematopoietic progenitor cell (HSPC) induction, erythroid progenitor cell induction, and terminal erythroid maturation. Differentiation efficiency was confirmed by flow cytometry analysis, colony-forming unit (CFU) assay, morphological analyses, and qPCR-based gene expression analyses of HBB and HBG2. RESULTS: Both 2D and 3D differentiation protocols led to the induction of CD34+/CD43+ HSPCs. The 3D protocol showed good efficiency (>50%) and high productivity (45-fold) for HSPC induction and increased the frequency of BFU-E, CFU-E, CFU-GM, and CFU-GEMM colonies. We also produced CD71+/CD235a+ cells (>65%) with a 630-fold cell expansion relative to that at the beginning of the 3D protocol. After erythroid maturation, we observed 95% CD235a+/DRAQ5- enucleated cells, orthochromatic erythroblasts, and increased expression of fetal HBG2 compared to adult HBB. CONCLUSION: A robust 3D protocol for erythroid differentiation was identified using SCD-iPSCs and comparative analyses; however, the maturation step remains challenging and requires further development.


Sujet(s)
Drépanocytose , Cellules souches pluripotentes induites , Adulte , Humains , Différenciation cellulaire , Cellules souches hématopoïétiques , Précurseurs érythroïdes/métabolisme , Drépanocytose/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...