Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Haematologica ; 109(6): 1918-1932, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38105727

ABSTRACT

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.


Subject(s)
Anemia, Sickle Cell , Disease Models, Animal , Reperfusion Injury , Animals , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/complications , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/pharmacology , Humans , Male , Hypoxia/metabolism , Hypoxia/drug therapy
2.
Anal Bioanal Chem ; 416(22): 4941-4949, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39090264

ABSTRACT

Traumatic brain injury (TBI) is an alteration of brain function caused by a sudden transmission of an external force to the head. The biomechanical impact induces acute and chronic metabolic changes that highly contribute to injury evolution and outcome. TBI heterogeneity calls for approaches allowing the mapping of regional molecular and metabolic changes underpinning disease progression, with mass spectrometry imaging (MSI) as an efficient tool to study the spatial distribution of small metabolites. In this study, we applied an innovative targeted atmospheric pressure-MALDI mass spectrometry imaging (AP-MALDI MSI) approach, starting from an extensive list of metabolites, representative of different metabolic pathways, individually validated on the tissue under analysis with original standards using 2,5-dihydroxybenzoic acid (DHB), to characterize the impact of TBI on regional changes to small metabolites in the brain. Brains from sham and TBI mice obtained 21 days post-injury were analyzed to examine the spatial metabolic profile of small metabolites belonging to different metabolic pathways. By a whole brain analysis, we identified four metabolites (alanine, lysine, histidine, and inosine) with higher abundance in TBI than sham mice. Within the TBI group, lysine, histidine, and inosine were higher in the hemisphere ipsilateral to the biomechanical impact vs. the contralateral one. Images showed a major involvement of the ipsilateral thalamus characterized by the increase of arginine, lysine, histidine, and inosine and a significant reduction of glutamic acid, and N-acetylaspartic acid compared to the contralateral thalamus. These findings indicate high-resolution imaging mass spectrometry as a powerful tool to identify region-specific changes after a TBI to understand the metabolic changes underlying brain injury evolution.


Subject(s)
Brain Injuries, Traumatic , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Brain Injuries, Traumatic/metabolism , Animals , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Male , Mice, Inbred C57BL , Brain/metabolism
3.
Haematologica ; 108(5): 1335-1348, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36700398

ABSTRACT

Cardiomyopathy deeply affects quality of life and mortality of patients with b-thalassemia or with transfusion-dependent myelodysplastic syndromes. Recently, a link between Nrf2 activity and iron metabolism has been reported in liver ironoverload murine models. Here, we studied C57B6 mice as healthy control and nuclear erythroid factor-2 knockout (Nrf2-/-) male mice aged 4 and 12 months. Eleven-month-old wild-type and Nrf2-/- mice were fed with either standard diet or a diet containing 2.5% carbonyl-iron (iron overload [IO]) for 4 weeks. We show that Nrf2-/- mice develop an age-dependent cardiomyopathy, characterized by severe oxidation, degradation of SERCA2A and iron accumulation. This was associated with local hepcidin expression and increased serum non-transferrin-bound iron, which promotes maladaptive cardiac remodeling and interstitial fibrosis related to overactivation of the TGF-b pathway. When mice were exposed to IO diet, the absence of Nrf2 was paradoxically protective against further heart iron accumulation. Indeed, the combination of prolonged oxidation and the burst induced by IO diet resulted in activation of the unfolded protein response (UPR) system, which in turn promotes hepcidin expression independently from heart iron accumulation. In the heart of Hbbth3/+ mice, a model of b-thalassemia intermedia, despite the activation of Nrf2 pathway, we found severe protein oxidation, activation of UPR system and cardiac fibrosis independently from heart iron content. We describe the dual role of Nrf2 when aging is combined with IO and its novel interrelation with UPR system to ensure cell survival. We open a new perspective for early and intense treatment of cardiomyopathy in patients with b-thalassemia before the appearance of heart iron accumulation.


Subject(s)
Cardiomyopathies , Iron Overload , Thalassemia , Animals , Male , Mice , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Hepcidins , Iron/metabolism , Iron Overload/complications , Iron Overload/genetics , Iron Overload/metabolism , NF-E2-Related Factor 2/metabolism , Quality of Life , Thalassemia/complications , Thalassemia/genetics , Thalassemia/metabolism
4.
Blood ; 133(3): 252-265, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30404812

ABSTRACT

Resolvins (Rvs), endogenous lipid mediators, play a key role in the resolution of inflammation. Sickle cell disease (SCD), a genetic disorder of hemoglobin, is characterized by inflammatory and vaso-occlusive pathologies. We document altered proresolving events following hypoxia/reperfusion in humanized SCD mice. We demonstrate novel protective actions of 17R-resolvin D1 (17R-RvD1; 7S, 8R, 17R-trihydroxy-4Z, 9E, 11E, 13Z, 15E, 19Z-docosahexaenoic acid) in reducing ex vivo human SCD blood leukocyte recruitment by microvascular endothelial cells and in vivo neutrophil adhesion and transmigration. In SCD mice exposed to hypoxia/reoxygenation, oral administration of 17R -RvD1 reduces systemic/local inflammation and vascular dysfunction in lung and kidney. The mechanism of action of 17R-RvD1 involves (1) enhancement of SCD erythrocytes and polymorphonuclear leukocyte efferocytosis, (2) blunting of NF-κB activation, and (3) a reduction in inflammatory cytokines, vascular activation markers, and E-selectin expression. Thus, 17R-RvD1 might represent a new therapeutic strategy for the inflammatory vasculopathy of SCD.


Subject(s)
Anemia, Sickle Cell/complications , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Docosahexaenoic Acids/administration & dosage , Kidney Diseases/prevention & control , Pneumonia/prevention & control , Animals , Cytokines/metabolism , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/etiology , Pneumonia/pathology
6.
Haematologica ; 104(5): 919-928, 2019 05.
Article in English | MEDLINE | ID: mdl-30630982

ABSTRACT

Sickle cell disease is an autosomal recessive genetic red cell disorder with a worldwide distribution. Growing evidence suggests a possible involvement of complement activation in the severity of clinical complications of sickle cell disease. In this study we found activation of the alternative complement pathway with microvascular deposition of C5b-9 on skin biopsies from patients with sickle cell disease. There was also deposition of C3b on sickle red cell membranes, which is promoted locally by the exposure of phosphatidylserine. In addition, we showed for the first time a peculiar "stop-and-go" motion of sickle cell red blood cells on tumor factor-α-activated vascular endothelial surfaces. Using the C3b/iC3b binding plasma protein factor Has an inhibitor of C3b cell-cell interactions, we found that factor H and its domains 19-20 prevent the adhesion of sickle red cells to the endothelium, normalizing speed transition times of red cells. We documented that factor H acts by preventing the adhesion of sickle red cells to P-selectin and/or the Mac-1 receptor (CD11b/CD18), supporting the activation of the alternative pathway of complement as an additional mechanism in the pathogenesis of acute sickle cell related vaso-occlusive crises. Our data provide a rationale for further investigation of the potential contribution of factor H and other modulators of the alternative complement pathway with potential implications for the treatment of sickle cell disease.


Subject(s)
Anemia, Sickle Cell/pathology , Cell Adhesion , Complement Membrane Attack Complex/metabolism , Endothelium, Vascular/pathology , Erythrocytes, Abnormal/pathology , Erythrocytes/pathology , Adolescent , Adult , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/immunology , Anemia, Sickle Cell/metabolism , Case-Control Studies , Cell Communication , Cells, Cultured , Complement Factor H/genetics , Complement Factor H/metabolism , Complement Membrane Attack Complex/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Erythrocytes, Abnormal/immunology , Erythrocytes, Abnormal/metabolism , Female , Follow-Up Studies , Humans , Macrophage-1 Antigen/metabolism , Male , Middle Aged , P-Selectin/metabolism , Young Adult
7.
Am J Hematol ; 94(1): 10-20, 2019 01.
Article in English | MEDLINE | ID: mdl-30252956

ABSTRACT

The signaling cascade induced by the interaction of erythropoietin (EPO) with its receptor (EPO-R) is a key event of erythropoiesis. We present here data indicating that Fyn, a Src-family-kinase, participates in the EPO signaling-pathway, since Fyn-/- mice exhibit reduced Tyr-phosphorylation of EPO-R and decreased STAT5-activity. The importance of Fyn in erythropoiesis is also supported by the blunted responsiveness of Fyn-/- mice to stress erythropoiesis. Fyn-/- mouse erythroblasts adapt to reactive oxygen species (ROS) by activating the redox-related-transcription-factor Nrf2. However, since Fyn is a physiologic repressor of Nrf2, absence of Fyn resulted in persistent-activation of Nrf2 and accumulation of nonfunctional proteins. ROS-induced over-activation of Jak2-Akt-mTOR-pathway and repression of autophagy with perturbation of lysosomal-clearance were also noted. Treatment with Rapamycin, a mTOR-inhibitor and autophagy activator, ameliorates Fyn-/- mouse baseline erythropoiesis and erythropoietic response to oxidative-stress. These findings identify a novel multimodal action of Fyn in the regulation of normal and stress erythropoiesis.


Subject(s)
Erythropoiesis/physiology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Autophagy , Doxorubicin/toxicity , Erythroblasts/enzymology , Erythropoiesis/drug effects , Erythropoiesis/genetics , Female , Janus Kinase 2/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Phenylhydrazines/toxicity , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Reactive Oxygen Species , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
8.
Blood ; 128(25): 2976-2987, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27742708

ABSTRACT

Chorea-acanthocytosis is one of the hereditary neurodegenerative disorders known as the neuroacanthocytoses. Chorea-acanthocytosis is characterized by circulating acanthocytes deficient in chorein, a protein of unknown function. We report here for the first time that chorea-acanthocytosis red cells are characterized by impaired autophagy, with cytoplasmic accumulation of active Lyn and of autophagy-related proteins Ulk1 and Atg7. In chorea-acanthocytosis erythrocytes, active Lyn is sequestered by HSP90-70 to form high-molecular-weight complexes that stabilize and protect Lyn from its proteasomal degradation, contributing to toxic Lyn accumulation. An interplay between accumulation of active Lyn and autophagy was found in chorea-acanthocytosis based on Lyn coimmunoprecipitation with Ulk1 and Atg7 and on the presence of Ulk1 in Lyn-containing high-molecular-weight complexes. In addition, chorein associated with Atg7 in healthy but not in chorea-acanthocytosis erythrocytes. Electron microscopy detected multivesicular bodies and membrane remnants only in circulating chorea-acanthocytosis red cells. In addition, reticulocyte-enriched chorea-acanthocytosis red cell fractions exhibited delayed clearance of mitochondria and lysosomes, further supporting the impairment of authophagic flux. Because autophagy is also important in erythropoiesis, we studied in vitro CD34+-derived erythroid precursors. In chorea-acanthocytosis, we found (1) dyserythropoiesis; (2) increased active Lyn; (3) accumulation of a marker of autophagic flux and autolysososme degradation; (4) accumlation of Lamp1, a lysosmal membrane protein, and LAMP1-positive aggregates; and (5) reduced clearance of lysosomes and mitochondria. Our results uncover in chorea-acanthocytosis erythroid cells an association between accumulation of active Lyn and impaired autophagy, suggesting a link between chorein and autophagic vesicle trafficking in erythroid maturation.


Subject(s)
Autophagy , Erythroid Cells/pathology , Neuroacanthocytosis/pathology , Adult , Anion Exchange Protein 1, Erythrocyte/metabolism , Autophagy/drug effects , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Benzoquinones/pharmacology , Bortezomib/pharmacology , Cell Differentiation/drug effects , Cytosol/drug effects , Cytosol/metabolism , Demography , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/pathology , Erythrocytes/ultrastructure , Erythroid Cells/drug effects , Erythroid Cells/ultrastructure , Erythropoiesis/drug effects , Female , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Male , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Weight , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , src-Family Kinases/metabolism
10.
Bioorg Med Chem Lett ; 28(17): 2816-2826, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30122223

ABSTRACT

Cancer still represents a "nightmare" worldwide, causing annually millions of victims. Several antiproliferative molecules are currently used as drugs market and offer a pharmaceutical opportunity for attenuating and treating tumor manifestations. In this context, natural sources have a relevant role, since they provide the 60% of currently-used anticancer agents. Among the numerous natural products, acting via different mechanisms of action, microtubule-targeting agents (MTAs) have a high therapeutic potential, since they disrupt the abnormal cancer cell growth, interfering with the continuous mitotic division. Vinca alkaloids (VAs) are the earliest developed MTAs and approved for clinical use (Vincristine, Vinblastine, Vinorelbine, Vindesine, and Vinflunine) as agents in the treatment of hematological and lymphatic neoplasms. Here, we review the state-of-art of VAs, discussing their mechanism of action and pharmacokinetic properties and highlighting their therapeutic relevance and toxicological profile. Additionally, we briefly disclosed the technological approaches faced so far to ameliorate the pharmacological properties, as well as to avoid the drug resistance. Lastly, we introduced the recent advances in the discovery of new derivatives.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Vinca Alkaloids/therapeutic use , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Humans , Neoplasms/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Vinca Alkaloids/chemistry
11.
Blood ; 126(20): 2320-8, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26330244

ABSTRACT

Sickle cell disease (SCD) is a worldwide distributed hereditary red cell disorder, characterized by severe organ complication. Sickle bone disease (SBD) affects a large part of the SCD patient population, and its pathogenesis has been only partially investigated. Here, we studied bone homeostasis in a humanized mouse model for SCD. Under normoxia, SCD mice display bone loss and bone impairment, with increased osteoclast and reduced osteoblast activity. Hypoxia/reperfusion (H/R) stress, mimicking acute vaso-occlusive crises (VOCs), increased bone turnover, osteoclast activity (RankL), and osteoclast recruitment (Rank) with upregulation of IL-6 as proresorptive cytokine. This was associated with further suppression of osteogenic lineage (Runx2, Sparc). To interfere with the development of SBD, zoledronic acid (Zol), a potent inhibitor of osteoclast activity/osteoclastogenesis and promoter of osteogenic lineage, was used in H/R-exposed mice. Zol markedly inhibited osteoclast activity and recruitment, promoting osteogenic lineage. The recurrent H/R stress further worsened bone structure, increased bone turnover, depressed osteoblastogenesis (Runx2, Sparc), and increased both osteoclast activity (RankL, Cathepsin k) and osteoclast recruitment (Rank) in SCD mice compared with either normoxic or single-H/R-episode SCD mice. Zol used before recurrent VOCs prevented bone impairment and promoted osteogenic lineage. Our findings support the view that SBD is related to osteoblast impairment, and increased osteoclast activity resulted from local hypoxia, oxidative stress, and the release of proresorptive cytokine such as IL-6. Zol might act on both the osteoclast and osteoblast compartments as multimodal therapy to prevent SBD.


Subject(s)
Anemia, Sickle Cell/metabolism , Bone Diseases/metabolism , Osteoclasts/metabolism , Reperfusion Injury/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Bone Diseases/genetics , Bone Diseases/pathology , Cathepsin K/genetics , Cathepsin K/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Diphosphonates/pharmacology , Disease Models, Animal , Humans , Imidazoles/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Transgenic , Osteoclasts/pathology , Osteonectin/genetics , Osteonectin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Zoledronic Acid
12.
Liver Int ; 36(1): 49-58, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26104535

ABSTRACT

BACKGROUND & AIMS: The addition of protease inhibitors, boceprevir (BOC) or telaprevir (TRV), to peg-interferon and ribavirin (PR) increases the incidence of anaemia in patients with chronic hepatitis C virus (HCV) infection. Although genetic variants in inosine triphosphatase (ITPA) gene have been linked to the haemolytic anaemia induced by PR, the mechanism sustaining severe anaemia during triple therapy is still unknown. This study aims to elucidate the molecular mechanisms underlying anaemia in chronic HCV patients with combined therapy. METHODS: We studied 59 patients with chronic HCV genotype-1: 29 treated with TRV/PR and 30 with BOC/PR. We evaluated biochemical and haematological parameters, red cell index at baseline, 4, 12, 16 and 24 weeks of treatment; in a subgroup, we performed functional studies: osmotic fragility, red cell membrane protein separation, mass spectrometry analysis, quantification of erythroid microparticles release. IL28B and ITPA polymorphisms were also evaluated. RESULTS: We found early acute normochromic normocytic haemolytic anaemia (4-8 weeks) followed by a late macrocytic hypo-regenerative anaemia with inappropriate low reticulocyte count (12-24 weeks). Studies on red cells revealed: (i) presence of spherocytes; (ii) increased osmotic fragility; (iii) abnormalities in red cell membrane protein composition; (iv) reduced membrane-cytoskeleton stability; (v) increased release of erythroid microparticles. ITPA polymorphisms impacted only the early phase of anaemia. CONCLUSIONS: The bimodal pattern of anaemia in chronic HCV patients on triple therapy might be because of acquired spherocytic-like anaemia in the early phase, followed by hyporegenerative anaemia, most likely related to the combined effects of PR and TRV or BOC on erythropoiesis.


Subject(s)
Anemia , Erythropoiesis/drug effects , Hepatitis C, Chronic , Oligopeptides , Proline/analogs & derivatives , Adult , Aged , Anemia/blood , Anemia/chemically induced , Anemia/diagnosis , Anemia/genetics , Anemia/physiopathology , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Drug Monitoring/methods , Drug Therapy, Combination , Erythrocyte Indices , Erythrocyte Membrane , Female , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/drug therapy , Humans , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Male , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Polymorphism, Single Nucleotide , Proline/administration & dosage , Proline/adverse effects , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Pyrophosphatases/genetics , Ribavirin/administration & dosage , Ribavirin/adverse effects , Treatment Outcome
13.
Haematologica ; 100(7): 870-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934765

ABSTRACT

The anemia of sickle cell disease is associated with a severe inflammatory vasculopathy and endothelial dysfunction, which leads to painful and life-threatening clinical complications. Growing evidence supports the anti-inflammatory properties of ω-3 fatty acids in clinical models of endothelial dysfunction. Promising but limited studies show potential therapeutic effects of ω-3 fatty acid supplementation in sickle cell disease. Here, we treated humanized healthy and sickle cell mice for 6 weeks with ω-3 fatty acid diet (fish-oil diet). We found that a ω-3 fatty acid diet: (i) normalizes red cell membrane ω-6/ω-3 ratio; (ii) reduces neutrophil count; (iii) decreases endothelial activation by targeting endothelin-1 and (iv) improves left ventricular outflow tract dimensions. In a hypoxia-reoxygenation model of acute vaso-occlusive crisis, a ω-3 fatty acid diet reduced systemic and local inflammation and protected against sickle cell-related end-organ injury. Using isolated aortas from sickle cell mice exposed to hypoxia-reoxygenation, we demonstrated a direct impact of a ω-3 fatty acid diet on vascular activation, inflammation, and anti-oxidant systems. Our data provide the rationale for ω-3 dietary supplementation as a therapeutic intervention to reduce vascular dysfunction in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/diet therapy , Anti-Inflammatory Agents/pharmacology , Blood Vessels/drug effects , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Disease Models, Animal , Endothelin-1/antagonists & inhibitors , Endothelin-1/biosynthesis , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/pathology , Humans , Hypoxia/diet therapy , Hypoxia/metabolism , Hypoxia/pathology , Mice , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Oxygen/adverse effects
14.
Haematologica ; 99(2): 267-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23975182

ABSTRACT

Resveratrol, a polyphenolic-stilbene, has received increased attention in the last decade due to its wide range of biological activities. Beta(ß)-thalassemias are inherited red cell disorders, found worldwide, characterized by ineffective erythropoiesis and red cell oxidative damage with reduced survival. We evaluated the effects of low-dose-resveratrol (5 µM) on in vitro human erythroid differentiation of CD34(+) from normal and ß-thalassemic subjects. We found that resveratrol induces accelerated erythroid-maturation, resulting in the reduction of colony-forming units of erythroid cells and increased intermediate and late erythroblasts. In sorted colony-forming units of erythroid cells resveratrol activates Forkhead-box-class-O3, decreases Akt activity and up-regulates anti-oxidant enzymes as catalase. In an in vivo murine model for ß-thalassemia, resveratrol (2.4 mg/kg) reduces ineffective erythropoiesis, increases hemoglobin levels, reduces reticulocyte count and ameliorates red cell survival. In both wild-type and ß-thalassemic mice, resveratrol up-regulates scavenging enzymes such as catalase and peroxiredoxin-2 through Forkhead-box-class-O3 activation. These data indicate that resveratrol inhibits Akt resulting in FoxO3 activation with upregulation of cytoprotective systems enabling the pathological erythroid precursors to resist the oxidative damage and continue to differentiate. Our data suggest that the dual effect of resveratrol on erythropoiesis through activation of FoxO3 transcriptional factor combined with the amelioration of oxidative stress in circulating red cells may be considered as a potential novel therapeutic strategy in treating ß-thalassemia.


Subject(s)
Enzyme Inhibitors/pharmacology , Erythrocytes/metabolism , Erythropoiesis/drug effects , Forkhead Transcription Factors/metabolism , Stilbenes/pharmacology , beta-Thalassemia/metabolism , Animals , Catalase/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Erythrocytes/pathology , Forkhead Box Protein O3 , Humans , Male , Mice , Peroxiredoxins/metabolism , Resveratrol , beta-Thalassemia/drug therapy , beta-Thalassemia/pathology
15.
FASEB J ; 27(2): 750-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085996

ABSTRACT

Sickle cell disease (SCD) is a globally distributed hereditary red blood cell (RBC) disorder. One of the hallmarks of SCD is the presence of circulating dense RBCs, which are important in SCD-related clinical manifestations. In human dense sickle cells, we found reduced calpastatin activity and protein expression compared to either healthy RBCs or unfractionated sickle cells, suggesting an imbalance between activator and inhibitor of calpain-1 in favor of activator in dense sickle cells. Calpain-1 is a nonlysosomal cysteine proteinase that modulates multiple cell functions through the selective cleavage of proteins. To investigate the relevance of this observation in vivo, we evaluated the effects of the orally active inhibitor of calpain-1, BDA-410 (30 mg/kg/d), on RBCs from SAD mice, a mouse model for SCD. In SAD mice, BDA-410 improved RBC morphology, reduced RBC density (D(20); from 1106 ± 0.001 to 1100 ± 0.001 g/ml; P<0.05) and increased RBC-K(+) content (from 364 ± 10 to 429 ± 12.3 mmol/kg Hb; P<0.05), markedly reduced the activity of the Ca(2+)-activated K(+)channel (Gardos channel), and decreased membrane association of peroxiredoxin-2. The inhibitory effect of calphostin C, a specific inhibitor of protein kinase C (PKC), on the Gardos channel was eliminated after BDA-410 treatment, which suggests that calpain-1 inhibition affects the PKC-dependent fraction of the Gardos channel. BDA-410 prevented hypoxia-induced RBC dehydration and K(+) loss in SAD mice. These data suggest a potential role of BDA-410 as a novel therapeutic agent for treatment of SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Calpain/antagonists & inhibitors , Erythrocytes, Abnormal/drug effects , Erythrocytes, Abnormal/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/blood , Anemia, Sickle Cell/genetics , Animals , Calcium-Binding Proteins/blood , Calpain/blood , Cysteine Proteinase Inhibitors/pharmacology , Dehydration/blood , Dehydration/prevention & control , Disease Models, Animal , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Mice , Mice, Mutant Strains , Mice, Transgenic , Sulfonamides/pharmacology
16.
Antioxidants (Basel) ; 13(4)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38671902

ABSTRACT

Aging is characterized by increased oxidation and reduced efficiency of cytoprotective mechanisms. Nuclear factor erythroid-2-related factor (Nrf2) is a key transcription factor, controlling the expression of multiple antioxidant proteins. Here, we show that Nrf2-/- mice displayed an age-dependent anemia, due to the combined contributions of reduced red cell lifespan and ineffective erythropoiesis, suggesting a role of Nrf2 in erythroid biology during aging. Mechanistically, we found that the expression of antioxidants during aging is mediated by activation of Nrf2 function by peroxiredoxin-2. The absence of Nrf2 resulted in persistent oxidation and overactivation of adaptive systems such as the unfolded protein response (UPR) system and autophagy in Nrf2-/- mouse erythroblasts. As Nrf2 is involved in the expression of autophagy-related proteins such as autophagy-related protein (Atg) 4-5 and p62, we found impairment of late phase of autophagy in Nrf2-/- mouse erythroblasts. The overactivation of the UPR system and impaired autophagy drove apoptosis of Nrf2-/- mouse erythroblasts via caspase-3 activation. As a proof of concept for the role of oxidation, we treated Nrf2-/- mice with astaxanthin, an antioxidant, in the form of poly (lactic-co-glycolic acid) (PLGA)-loaded nanoparticles (ATS-NPs) to improve its bioavailability. ATS-NPs ameliorated the age-dependent anemia and decreased ineffective erythropoiesis in Nrf2-/- mice. In summary, we propose that Nrf2 plays a key role in limiting age-related oxidation, ensuring erythroid maturation and growth during aging.

17.
Blood ; 118(20): 5652-63, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-21951684

ABSTRACT

Acanthocytic RBCs are a peculiar diagnostic feature of chorea-acanthocytosis (ChAc), a rare autosomal recessive neurodegenerative disorder. Although recent years have witnessed some progress in the molecular characterization of ChAc, the mechanism(s) responsible for generation of acanthocytes in ChAc is largely unknown. As the membrane protein composition of ChAc RBCs is similar to that of normal RBCs, we evaluated the tyrosine (Tyr)-phosphorylation profile of RBCs using comparative proteomics. Increased Tyr phosphorylation state of several membrane proteins, including band 3, ß-spectrin, and adducin, was noted in ChAc RBCs. In particular, band 3 was highly phosphorylated on the Tyr-904 residue, a functional target of Lyn, but not on Tyr-8, a functional target of Syk. In ChAc RBCs, band 3 Tyr phosphorylation by Lyn was independent of the canonical Syk-mediated pathway. The ChAc-associated alterations in RBC membrane protein organization appear to be the result of increased Tyr phosphorylation leading to altered linkage of band 3 to the junctional complexes involved in anchoring the membrane to the cytoskeleton as supported by coimmunoprecipitation of ß-adducin with band 3 only in ChAc RBC-membrane treated with the Lyn-inhibitor PP2. We propose this altered association between membrane skeleton and membrane proteins as novel mechanism in the generation of acanthocytes in ChAc.


Subject(s)
Acanthocytes/enzymology , Erythrocyte Membrane/enzymology , Neuroacanthocytosis/metabolism , src-Family Kinases/metabolism , Acanthocytes/pathology , Adult , Anion Exchange Protein 1, Erythrocyte/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Enzyme Activation/physiology , Erythrocyte Membrane/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Neuroacanthocytosis/pathology , Phosphorylation/physiology , Protein-Tyrosine Kinases/metabolism , Proteomics , Syk Kinase , Tyrosine/metabolism
18.
Hemasphere ; 7(3): e848, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36874380

ABSTRACT

Drug repurposing is a valuable strategy for rare diseases. Sickle cell disease (SCD) is a rare hereditary hemolytic anemia accompanied by acute and chronic painful episodes, most often in the context of vaso-occlusive crisis (VOC). Although progress in the knowledge of pathophysiology of SCD have allowed the development of new therapeutic options, a large fraction of patients still exhibits unmet therapeutic needs, with persistence of VOCs and chronic disease progression. Here, we show that imatinib, an oral tyrosine kinase inhibitor developed for the treatment of chronic myelogenous leukemia, acts as multimodal therapy targeting signal transduction pathways involved in the pathogenesis of both anemia and inflammatory vasculopathy of humanized murine model for SCD. In addition, imatinib inhibits the platelet-derived growth factor-B-dependent pathway, interfering with the profibrotic response to hypoxia/reperfusion injury, used to mimic acute VOCs. Our data indicate that imatinib might be considered as possible new therapeutic tool for chronic treatment of SCD.

19.
Haematologica ; 96(1): 24-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20851863

ABSTRACT

BACKGROUND: Sickle cell disease, a genetic red cell disorder inherited in an autosomal recessive manner, occurs throughout the world. Hepatic dysfunction and liver damage may be present in sickle cell disease, but the pathogenesis of these conditions is only partially understood. DESIGN AND METHODS: Transgenic mice with sickle cell disease (SAD mice) and wild-type mice were exposed to an ischemic/reperfusion stress. The following parameters were evaluated: hematologic profile, transaminase and bilirubin levels, liver histopathology, and mRNA levels of nuclear factor-κB p65, endothelial nitric oxide synthase, inducible nitric oxide synthase, heme oxygenase-1 and phosphodiesterase-1, -2, -3, and -4 genes in hepatocytes obtained by laser-capture microdissection. Immunoblotting was used to analyze the expression of the following proteins: nuclear factor-κB p65 and phospho-nuclear factor-κB p65, heme oxygenase-1, biliverdin reductase, heat shock protein-70, heat shock protein-27 and peroxiredoxin-6. A subgroup of SAD mice was treated with the phosphodiesterase-4 inhibitor rolipram (30 mg/Kg/day by gavage) during the ischemic/reperfusion protocol. RESULTS: In SAD mice the ischemic/reperfusion stress induced liver damage compatible with sickle cell disease hepatopathy, which was associated with: (i) lack of hypoxia-induced nuclear factor-κB p65 activation; (ii) imbalance in the endothelial/inducible nitric oxide synthase response to ischemic/reperfusion stress; (iii) lack of hypoxia-induced increased expression of heme oxygenase-1/biliverdin reductase paralleled by a compensatory increased expression of heat shock proteins 70 and 27 and peroxiredoxin-6; and (iv) up-regulation of the phosphodiesterase-1, -2, -3, and -4 genes. In SAD mice the phosphodiesterase-4 inhibitor rolipram attenuated the ischemic/reperfusion-related microcirculatory dysfunction, reduced the inflammatory cell infiltration and induced the heme oxygenase-1/biliverdin reductase cytoprotective systems. CONCLUSIONS: In SAD mice, sickle cell hepatopathy is associated with perturbed nuclear factor-κB p65 signaling with an imbalance of endothelial/inducible nitric oxide synthase levels, lack of heme oxygenase-1/biliverdin reductase expression and up-regulation of two novel cytoprotective systems: heat shock protein-27 and peroxiredoxin-6.


Subject(s)
Anemia, Sickle Cell/etiology , Cytoprotection , Liver Diseases/etiology , Liver Diseases/pathology , Reperfusion Injury/complications , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Animals , Blotting, Western , Cells, Cultured , Female , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , RNA, Messenger/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction
20.
Antioxidants (Basel) ; 11(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35052580

ABSTRACT

The peroxiredoxins (PRXs) constitute a ubiquitous antioxidant. Growing evidence in neurodegenerative disorders such as Parkinson's disease (PD) or Alzheimer's disease (AD) has highlighted a crucial role for PRXs against neuro-oxidation. Chorea-acanthocytosis/Vps13A disease (ChAc) is a devastating, life-shortening disorder characterized by acanthocytosis, neurodegeneration and abnormal proteostasis. We recently developed a Vps13a-/- ChAc-mouse model, showing acanthocytosis, neurodegeneration and neuroinflammation which could be restored by LYN inactivation. Here, we show in our Vps13a-/- mice protein oxidation, NRF2 activation and upregulation of downstream cytoprotective systems NQO1, SRXN1 and TRXR in basal ganglia. This was associated with upregulation of PRX2/5 expression compared to wild-type mice. PRX2 expression was age-dependent in both mouse strains, whereas only Vps13a-/- PRX5 expression was increased independent of age. LYN deficiency or nilotinib-mediated LYN inhibition improved autophagy in Vps13a-/- mice. In Vps13a-/-; Lyn-/- basal ganglia, absence of LYN resulted in reduced NRF2 activation and down-regulated expression of PRX2/5, SRXN1 and TRXR. Nilotinib treatment of Vps13a-/- mice reduced basal ganglia oxidation, and plasma PRX5 levels, suggesting plasma PRX5 as a possible ChAc biomarker. Our data support initiation of therapeutic Lyn inhibition as promptly as possible after ChAc diagnosis to minimize development of irreversible neuronal damage during otherwise inevitable ChAc progression.

SELECTION OF CITATIONS
SEARCH DETAIL