Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.416
Filter
1.
J Cell Mol Med ; 28(9): e18308, 2024 May.
Article in English | MEDLINE | ID: mdl-38683131

ABSTRACT

Destruction of erythropoiesis process leads to various diseases, including thrombocytopenia, anaemia, and leukaemia. miR-429-CT10 regulation of kinase-like (CRKL) axis involved in development, progression and metastasis of cancers. However, the exact role of miR-429-CRKL axis in leukaemic cell differentiation are still unknown. The current work aimed to uncover the effect of miR-429-CRKL axis on erythropoiesis. In the present study, CRKL upregulation was negatively correlated with miR-429 downregulation in both chronic myeloid leukaemia (CML) patient and CR patient samples. Moreover, CRKL expression level was significantly decreased while miR-429 expression level was increased during the erythroid differentiation of K562 cells following hemin treatment. Functional investigations revealed that overexpression and knockdown of CRKL was remarkably effective in suppressing and promoting hemin-induced erythroid differentiation of K562 cells, whereas, miR-429 exhibited opposite effects to CRKL. Mechanistically, miR-429 regulates erythroid differentiation of K562 cells by downregulating CRKL via selectively targeting CRKL-3'-untranslated region (UTR) through Raf/MEK/ERK pathway. Conversely, CRKII had no effect on erythroid differentiation of K562 cells. Taken together, our data demonstrated that CRKL (but not CRKII) and miR-429 contribute to development, progression and erythropoiesis of CML, miR-429-CRKL axis regulates erythropoiesis of K562 cells via Raf/MEK/ERK pathway, providing novel insights into effective diagnosis and therapy for CML patients.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Erythroid Cells , Hemin , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MicroRNAs , Proto-Oncogene Proteins c-crk , Humans , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation/drug effects , Erythroid Cells/metabolism , Erythroid Cells/drug effects , Erythroid Cells/pathology , Erythroid Cells/cytology , Erythropoiesis/genetics , Erythropoiesis/drug effects , Gene Expression Regulation, Leukemic/drug effects , Hemin/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , MAP Kinase Signaling System/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Proto-Oncogene Proteins c-crk/genetics
2.
Nefrologia (Engl Ed) ; 44(2): 165-172, 2024.
Article in English | MEDLINE | ID: mdl-38604895

ABSTRACT

Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have demonstrated cardiovascular and renal benefits in patients with type 2 diabetes mellitus, heart failure, or chronic kidney disease. Since the first studies with these drugs, an initial increase in hemoglobin/hematocrit levels was observed, which was attributed to an increase in hemoconcentration associated with its diuretic effect, although it was early appearent that these drugs increased erythropoietin levels and erythropoiesis, and improved iron metabolism. Mediation studies found that the increase in hemoglobin was strongly associated with the cardiorenal benefits of these drugs. In this review, we discuss the mechanisms for improving erythropoiesis and the implication of the increase in hemoglobin on the cardiorenal prognostic benefit of these drugs.


Subject(s)
Anemia , Diabetes Mellitus, Type 2 , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Humans , Anemia/drug therapy , Anemia/etiology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Erythropoiesis/drug effects , Renal Insufficiency, Chronic/complications , Heart Failure/drug therapy , Hemoglobins/analysis , Erythropoietin/therapeutic use
3.
Br J Haematol ; 204(5): 2077-2085, 2024 May.
Article in English | MEDLINE | ID: mdl-38462764

ABSTRACT

Diamond-Blackfan anaemia (DBA) is a rare, inherited bone marrow failure syndrome with a ribosomal defect causing slowed globin chain production with normal haem synthesis, causing an overabundance of reactive iron/haem and erythroid-specific cellular toxicity. Eltrombopag, a non-peptide thrombopoietin receptor agonist, is a potent intracellular iron chelator and induced a robust durable response in an RPS19-mutated DBA patient on another trial. We hypothesized eltrombopag would improve RBC production in DBA patients. We conducted a single-centre, single-arm pilot study (NCT04269889) assessing safety and erythroid response of 6 months of daily, fixed-dose eltrombopag for DBA patients. Fifteen transfusion-dependent (every 3-5 weeks) patients (median age 18 [range 2-56]) were treated. One responder had sustained haemoglobin improvement and >50% reduction in RBC transfusion frequency. Of note, 7/15 (41%) patients required dose reductions or sustained discontinuation of eltrombopag due to asymptomatic thrombocytosis. Despite the low response rate, eltrombopag has now improved erythropoiesis in several patients with DBA with a favourable safety profile. Dosing restrictions due to thrombocytosis may cause insufficient iron chelation to decrease haem production and improve anaemia in most patients. Future work will focus on erythropoiesis dynamics in patients and use of haem synthesis inhibitors without an impact on other haematopoietic lineages.


Subject(s)
Anemia, Diamond-Blackfan , Benzoates , Hydrazines , Pyrazoles , Humans , Anemia, Diamond-Blackfan/drug therapy , Pyrazoles/therapeutic use , Hydrazines/therapeutic use , Hydrazines/administration & dosage , Hydrazines/adverse effects , Benzoates/therapeutic use , Benzoates/administration & dosage , Benzoates/adverse effects , Adult , Male , Female , Child , Adolescent , Middle Aged , Young Adult , Child, Preschool , Pilot Projects , Treatment Outcome , Receptors, Thrombopoietin/agonists , Recurrence , Erythropoiesis/drug effects
4.
Blood Adv ; 8(10): 2433-2441, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38330179

ABSTRACT

ABSTRACT: Pyruvate kinase (PK) deficiency is a rare, hereditary disease characterized by chronic hemolytic anemia. Iron overload is a common complication regardless of age, genotype, or transfusion history. Mitapivat, an oral, allosteric PK activator, improves anemia and hemolysis in adult patients with PK deficiency. Mitapivat's impact on iron overload and ineffective erythropoiesis was evaluated in adults with PK deficiency who were not regularly transfused in the phase 3 ACTIVATE trial and long-term extension (LTE) (#NCT03548220/#NCT03853798). Patients in the LTE received mitapivat throughout ACTIVATE/LTE (baseline to week 96; mitapivat-to-mitapivat [M/M] arm) or switched from placebo (baseline to week 24) to mitapivat (week 24 to week 96; placebo-to-mitapivat [P/M] arm). Changes from baseline in markers of iron overload and erythropoiesis were assessed to week 96. Improvements in hepcidin (mean, 4770.0 ng/L; 95% confidence interval [CI], -1532.3 to 11 072.3), erythroferrone (mean, -9834.9 ng/L; 95% CI, -14 328.4 to -5341.3), soluble transferrin receptor (mean, -56.0 nmol/L; 95% CI, -84.8 to -27.2), and erythropoietin (mean, -32.85 IU/L; 95% CI, -54.65 to -11.06) were observed in the M/M arm (n = 40) from baseline to week 24, sustained to week 96. No improvements were observed in the P/M arm (n = 40) to week 24; however, upon transitioning to mitapivat, improvements similar to those observed in the M/M arm were seen. Mean changes from baseline in liver iron concentration by magnetic resonance imaging at week 96 in the M/M arm and the P/M arm were -2.0 mg Fe/g dry weight (dw; 95% CI, -4.8 to -0.8) and -1.8 mg Fe/g dw (95% CI, -4.4 to 0.80), respectively. Mitapivat is the first disease-modifying pharmacotherapy shown to have beneficial effects on iron overload and ineffective erythropoiesis in patients with PK deficiency. This trial was registered at www.ClinicalTrials.gov as #NCT03548220 (ACTIVATE) and #NCT03853798 (LTE).


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Erythropoiesis , Iron Overload , Pyruvate Kinase , Pyruvate Metabolism, Inborn Errors , Humans , Iron Overload/etiology , Iron Overload/drug therapy , Erythropoiesis/drug effects , Adult , Pyruvate Kinase/deficiency , Male , Female , Middle Aged , Young Adult , Alanine/therapeutic use , Alanine/analogs & derivatives , Piperazines , Quinolines
5.
Rev. clín. esp. (Ed. impr.) ; 223(3): 181-187, mar. 2023.
Article in Spanish | IBECS | ID: ibc-217182

ABSTRACT

Entre las sustancias prohibidas por la Agencia Mundial Antidopaje, en el apartado S2 se clasifican como prohibidas, tanto en competición como fuera de competición, las «hormonas peptídicas, factores de crecimiento y sustancias relacionadas y miméticos». En este trabajo realizamos una revisión de: la hormona de crecimiento y sus péptidos liberadores; el factor de crecimiento similar a la insulina tipo 1 como principal factor de crecimiento; la insulina, y la eritropoyetina y otros agentes que afectan la eritropoyesis. En esta revisión analizamos la prevalencia de uso en deportistas profesionales y clientes de gimnasios; las formas de uso, dosis, efectos ergogénicos y sobre el rendimiento físico, así como los efectos secundarios y métodos de detección antidopaje (AU)


Among the substances prohibited by the World Anti-Doping Agency, “peptide hormones, growth factors, related substances, and mimetics” are classified as prohibited both in- and out-of-competition in section S2. This work reviews growth hormone and its releasing peptides, insulin-like growth factor 1 as the main growth factor, insulin, and erythropoietin and other agents that affect erythropoiesis. This review analyzes the prevalence of use among professional athletes and gym clients, the forms of use, dosing, ergogenic effects and effects on physical performance, as well as side effects and anti-doping detection methods (AU)


Subject(s)
Humans , Doping in Sports , Epoetin Alfa/pharmacology , Erythropoietin/pharmacology , Human Growth Hormone/pharmacology , Erythropoiesis/drug effects , Insulin-Like Growth Factor I , Insulin, Regular, Human
6.
Int J Mol Sci ; 23(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-35163808

ABSTRACT

Diamond-Blackfan anaemia (DBA) is a red blood cell aplasia that in the majority of cases is associated with ribosomal protein (RP) aberrations. However, the mechanism by which this disorder leads to such a specific phenotype remains unclear. Even more elusive is the reason why non-specific agents such as glucocorticosteroids (GCs), also known as glucocorticoids, are an effective therapy for DBA. In this review, we (1) explore why GCs are successful in DBA treatment, (2) discuss the effect of GCs on erythropoiesis, and (3) summarise the GC impact on crucial pathways deregulated in DBA. Furthermore, we show that GCs do not regulate DBA erythropoiesis via a single mechanism but more likely via several interdependent pathways.


Subject(s)
Anemia, Diamond-Blackfan/drug therapy , Gene Regulatory Networks/drug effects , Glucocorticoids/therapeutic use , Anemia, Diamond-Blackfan/metabolism , Erythropoiesis/drug effects , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Humans , Treatment Outcome
7.
Exp Mol Pathol ; 124: 104740, 2022 02.
Article in English | MEDLINE | ID: mdl-34998864

ABSTRACT

Aloin, an anthraquinone glycoside, is one of other C-glycosides found in the leaf exudate of Aloe plant. Aloin possesses several biologic activities, including antitumor activity in vitro and in vivo. However, aloin treatment has shown iron deficiency anemia and erythropoiesis in vivo. The present study was undertaken to verify if iron supplementation could alleviate these perturbations, compared to doxorubicin, an anthracycline analog. Oral iron supplementation (20.56 mg elemental Fe/kg bw) to aloin-treated rats normalized red blood corpuscles count, hemoglobin concentration, and serum levels of total iron binding capacity and saturated transferrin, as well as hepatic iron content, hepcidin level, and mRNA expression of ferritin heavy chain (Ferr-H) and transferrin receptor-1 (TfR-1) genes. Although, serum hyperferremia, and leukocytosis were maintained, yet the spleen iron overload was substantially modulated. However, combined aloin and iron treatment increased iron storage levels in the heart and bone marrow, compared to aloin treatment per se. On other hand, oral iron supplementation to rats treated with doxorubicin (15 mg/kg bw) lessened the increase in the spleen iron content concomitantly with hepatic hepcidin level, rebound hepatic iron content to normal level, and by contrast augmented serum levels of iron and transferrin saturation. Also, activated Ferr-H mRNA expression and repressed TfR-1 mRNA expression were recorded, compared to doxorubicin treatment per se. Histopathological examination of the major body iron stores in rats supplemented with iron along with aloin or doxorubicin showed an increase in extramedullary hematopoiesis. In conclusion, iron supplementation restores the disturbances in iron homeostasis and erythropoiesis induced by aloin treatment.


Subject(s)
Anemia, Iron-Deficiency , Dietary Supplements , Emodin/analogs & derivatives , Iron , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Animals , Emodin/adverse effects , Emodin/pharmacology , Erythropoiesis/drug effects , Glycosides/adverse effects , Glycosides/pharmacology , Hepcidins/blood , Hepcidins/drug effects , Iron/metabolism , Iron/therapeutic use , Iron Deficiencies/drug therapy , Iron Deficiencies/metabolism , Liver/metabolism , Rats , Receptors, Transferrin/blood , Receptors, Transferrin/drug effects , Spleen/metabolism
9.
Sci Rep ; 11(1): 22121, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764389

ABSTRACT

Arsenic exposure poses numerous threats to human health. Our previous work in mice has shown that arsenic causes anemia by inhibiting erythropoiesis. However, the impacts of arsenic exposure on human erythropoiesis remain largely unclear. We report here that low-dose arsenic exposure inhibits the erythroid differentiation of human hematopoietic progenitor cells (HPCs). The impacts of arsenic (in the form of arsenite; As3+) on red blood cell (RBC) development was evaluated using a long-term culture of normal human bone marrow CD34+-HPCs stimulated in vitro to undergo erythropoiesis. Over the time course studied, we analyzed the expression of the cell surface antigens CD34, CD71 and CD235a, which are markers commonly used to monitor the progression of HPCs through the stages of erythropoiesis. Simultaneously, we measured hemoglobin content, which is an important criterion used clinically for diagnosing anemia. As compared to control, low-dose As3+ exposure (100 nM and 500 nM) inhibited the expansion of CD34+-HPCs over the time course investigated; decreased the number of committed erythroid progenitors (BFU-E and CFU-E) and erythroblast differentiation in the subsequent stages; and caused a reduction of hemoglobin content. These findings demonstrate that low-dose arsenic exposure impairs human erythropoiesis, likely by combined effects on various stages of RBC formation.


Subject(s)
Antigens, CD34/metabolism , Arsenites/adverse effects , Cell Differentiation/drug effects , Erythroid Precursor Cells/drug effects , Hematopoietic Stem Cells/drug effects , Hemoglobins/metabolism , Anemia/chemically induced , Anemia/metabolism , Antigens, CD/metabolism , Cells, Cultured , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Glycophorins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Receptors, Transferrin/metabolism
10.
Pediatr Blood Cancer ; 68(11): e29338, 2021 11.
Article in English | MEDLINE | ID: mdl-34520107

ABSTRACT

BACKGROUND: Ineffective erythropoiesis (IE) is the most prominent feature of transfusion-dependent beta-thalassemia (TDT), which leads to extramedullary hemopoiesis. The rejection rate in allogeneic hematopoietic stem cell transplantation (HSCT) is high in heavily transfused patients with TDT accompanied by prominent IE. Therefore, a pretransplantation treatment bridging to HSCT is often used to reduce allosensitization and IE. Ruxolitinib is a JAK-1/JAK-2 inhibitor and has showed its efficacy in suppressing IE and the immune system. A previously published study on RUX in adult patients with TDT has revealed that this treatment significantly reduces spleen size and is well tolerated. PROCEDURE: Ten patients (5-14 years old) with TDT and an enlarged spleen were enrolled. The dose of ruxolitinib was adjusted for age: for patients <11 years: 40-100 mg/m2 total daily dose and for patients >11 years: 20-30 mg/m2 total daily dose. HSCT was performed in 8 of 10 patients. RESULTS: After the first 3 months of ruxolitinib therapy, spleen volume decreased in 9 of 10 cases by 9.1%-67.5% (M = 35.4%) compared with the initial size (P = 0.003). The adverse events of ruxolitinib (infectious complications, moderate thrombocytopenia, and headache) were successfully managed by reducing the dose. The outcomes of HSCT were favorable in seven of eight cases. CONCLUSION: Ruxolitinib is promising as a short-term pre-HSCT treatment for pediatric patients with TDT and pronounced IE.


Subject(s)
Hematopoietic Stem Cell Transplantation , Nitriles/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , beta-Thalassemia , Adolescent , Child , Child, Preschool , Erythropoiesis/drug effects , Humans , beta-Thalassemia/therapy
11.
Am J Hematol ; 96(11): 1518-1531, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34347889

ABSTRACT

The treatment landscape for patients with ß-thalassemia is witnessing a swift evolution, yet several unmet needs continue to persist. Patients with transfusion-dependent ß-thalassemia (TDT) primarily rely on regular transfusion and iron chelation therapy, which can be associated with considerable treatment burden and cost. Patients with non-transfusion-dependent ß-thalassemia (NTDT) are also at risk of significant morbidity due to the underlying anemia and iron overload, but treatment options in this patient subgroup are limited. In this review, we provide updates on clinical trials of novel therapies targeting the underlying pathology in ß-thalassemia, including the α/non-α-globin chain imbalance, ineffective erythropoiesis, and iron dysregulation.


Subject(s)
beta-Thalassemia/therapy , Blood Transfusion , Clinical Trials as Topic , Drug Discovery , Erythropoiesis/drug effects , Humans , Iron/metabolism , Iron Chelating Agents/therapeutic use , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
12.
Life Sci ; 284: 119900, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34453946

ABSTRACT

ZMIZ1 is a transcriptional coactivator that is related to members of the protein inhibitor of activated STAT (PIAS) family. ZMIZ1 regulates the activity of various transcription factors including the androgen receptor, p53, and Smad3. ZMIZ1 also interacts with Notch1 and selectively regulates Notch1 target genes relevant for T cell development and leukemogenesis in mammals. Human ZMIZ1 is additionally characterized as a latitude-dependent autoimmune disease (LDAD) risk gene, as it is responsive to vitamin D and has been associated with at least eleven blood cell traits. To address the function of ZMIZ1 in fish, we introduced CRISPR/Cas9 mutations in the zmiz1a gene in zebrafish. We observed that inactivation of zmiz1a in developing zebrafish larvae results in lethality at 15 days post fertilization (dpf) and delayed erythroid maturation. Differential gene expression analysis indicated that 15 dpf zmiz1a-null larvae had altered expression of autophagy genes, and erythrocytes that lacked Zmiz1a function exhibited an accumulation of mitochondrial DNA. Furthermore, we observed that autophagy gene expression was dysregulated at earlier stages of development, which suggests the involvement of Zmiz1a in the regulation of autophagy genes beyond the process of red blood cell differentiation. Finally, we showed that the loss of Zmiz1a decreased the capacity of the embryos to respond to vitamin D, indicating additional participation of Zmiz1a as a mediator of vitamin D activity.


Subject(s)
Autophagy/genetics , Erythropoiesis/genetics , Gene Expression Regulation/drug effects , Mutation/genetics , Vitamin D/pharmacology , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Autophagy/drug effects , Base Sequence , Cell Differentiation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Nonmammalian/pathology , Erythrocytes/drug effects , Erythrocytes/pathology , Erythropoiesis/drug effects , Gene Dosage , Hemoglobins/metabolism , Inflammation/genetics , Transcriptome/genetics , Zebrafish/embryology , Zebrafish Proteins/metabolism
13.
Cells ; 10(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34440909

ABSTRACT

Human erythropoietin (EPO) is an N-linked glycoprotein consisting of 166 aa that is produced in the kidney during the adult life and acts both as a peptide hormone and hematopoietic growth factor (HGF), stimulating bone marrow erythropoiesis. EPO production is activated by hypoxia and is regulated via an oxygen-sensitive feedback loop. EPO acts via its homodimeric erythropoietin receptor (EPO-R) that increases cell survival and drives the terminal erythroid maturation of progenitors BFU-Es and CFU-Es to billions of mature RBCs. This pathway involves the activation of multiple erythroid transcription factors, such as GATA1, FOG1, TAL-1, EKLF and BCL11A, and leads to the overexpression of genes encoding enzymes involved in heme biosynthesis and the production of hemoglobin. The detection of a heterodimeric complex of EPO-R (consisting of one EPO-R chain and the CSF2RB ß-chain, CD131) in several tissues (brain, heart, skeletal muscle) explains the EPO pleotropic action as a protection factor for several cells, including the multipotent MSCs as well as cells modulating the innate and adaptive immunity arms. EPO induces the osteogenic and endothelial transdifferentiation of the multipotent MSCs via the activation of EPO-R signaling pathways, leading to bone remodeling, induction of angiogenesis and secretion of a large number of trophic factors (secretome). These diversely unique properties of EPO, taken together with its clinical use to treat anemias associated with chronic renal failure and other blood disorders, make it a valuable biologic agent in regenerative medicine for the treatment/cure of tissue de-regeneration disorders.


Subject(s)
Bone Remodeling/physiology , Cell Transdifferentiation/physiology , Endothelial Cells/cytology , Erythropoiesis/physiology , Erythropoietin/metabolism , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Bone Remodeling/drug effects , Cell Transdifferentiation/drug effects , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Humans , Receptors, Erythropoietin/metabolism , Regenerative Medicine/methods
14.
Drug Test Anal ; 13(10): 1791-1796, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34342157

ABSTRACT

Erythropoietin receptor agonists (ERAs) are drugs acting on the early erythropoietic stages developed to treat anemia and other erythropoiesis disease and are prohibited by the World Anti-Doping Agency (WADA). As an alternative to ERAs, a new drug, belonging to the transforming growth factor-b inhibitors family, was recently developed to treat diseases linked to ineffective erythropoiesis. This drug, named as Luspatercept (Reblozyl®), is acting on the later stages of erythropoiesis to promote erythrocytes. This drug might be used by cheating athletes either independently or in combination with ERAs. Indeed, it was shown that Luspatercept and recombinant erythropoietin (rEPO) can act synergistically to increase red blood cells production, potentially allowing the use of lower doses for an efficient effect. Our aim was to find a way to combine the detection of ERAs and Luspatercept without impacting the sensitivity and specificity of ERAs detection from the current techniques implemented in antidoping laboratories and to reduce the time of analysis and total sample volume needed. Magnetic beads coated with antibodies were preferred for IP of samples for its potential multiplexing. Then, the following steps of the method were selected considering that SAR/SDS-PAGE are the electrophoretic methods authorized for initial testing procedure by WADA and that biotinylated primary antibodies used for the immunodetection results in the best sensitivity and specificity and is time saving. The method developed in this work for the combined detection of agents affecting erythropoiesis (AAEs) showed specificity, sensitivity, and robustness and is easily and quickly implementable to all antidoping laboratories.


Subject(s)
Activin Receptors, Type II/analysis , Doping in Sports/prevention & control , Hematinics/analysis , Immunoglobulin Fc Fragments/analysis , Recombinant Fusion Proteins/analysis , Substance Abuse Detection/methods , Electrophoresis, Polyacrylamide Gel , Erythropoiesis/drug effects , Humans , Sensitivity and Specificity
15.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445665

ABSTRACT

Mast cell disease is an epigenetically and genetically determined disease entity with very diverse clinical manifestations in potentially every system and tissue due to inap pro priate release of variable subsets of mast cell mediators together with accumulation of either morphologically normal or altered mast cells. Easy bruising, excessive bleeding, and aberrancies of erythropoiesis can frequently be observed in patients with mast cell disease. A thorough history, including a family history, will guide the appropriate work-up, and laboratory evaluations may provide clues to diagnosis. In recent years, our understanding of the involvement of coagulation and anticoagulant pathways, the fibrinolytic system, and erythropoiesis in the pathophysiology of mast cell disease has increased considerably. This review summarizes current knowledge of the impact of the disturbed hemostatic and erythropoietic balance in patients with mast cell disease and describes options of treatment.


Subject(s)
Erythropoiesis/physiology , Hemostasis/physiology , Mastocytosis/blood , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Erythropoiesis/drug effects , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Hemostasis/drug effects , Heparin/pharmacology , Humans , Mastocytosis/immunology , Mastocytosis/physiopathology
16.
Toxicol Lett ; 350: 111-120, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34274428

ABSTRACT

Strong epidemiological evidence demonstrates an association between chronic arsenic exposure and anemia. We recently found that As+3 impairs erythropoiesis by disrupting the function of GATA-1; however the downstream pathways impacted by the loss of GATA-1 function have not been evaluated. Additionally, our previous findings indicate that the predominant arsenical in the bone marrow of mice exposed to As+3 in their drinking water for 30 days was MMA+3, but the impacts of this arsenical on erythorpoisis also remain largely unknown. The goal of this study was to address these critical knowledge gaps by evaluating the comparative effects of arsenite (As+3) and the As+3 metabolite, monomethyarsonous acid (MMA+3) on two critical regulatory pathways that control the differentiation and survival of early erythroid progenitor cells. We found that 500 nM As+3 and 100 and 500 nM MMA+3 suppress erythropoiesis by impairing the differentiation of early stage erythroid progenitors. The suppression of early erythroid progenitor cell development was attributed to combined effects on differentiation and survival pathways mediated by disruption of GATA-1 and STAT5. Our results show that As+3 primarily disrupted GATA-1 function; whereas, MMA+3 suppressed both GATA-1 and STAT5 activity. Collectively, these findings provide novel mechanistic insights into arsenic-induced dyserythropoiesis and suggest that MMA+3 may be more toxic than As+3 to early developing erythroid cells.


Subject(s)
Anemia/chemically induced , Arsenic/toxicity , Arsenites/toxicity , Cell Differentiation/drug effects , Cell Survival/drug effects , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Organometallic Compounds/toxicity , Animals , Humans , Mice , Models, Animal
17.
J Biol Chem ; 297(3): 100988, 2021 09.
Article in English | MEDLINE | ID: mdl-34298020

ABSTRACT

Nemo-like kinase (NLK) is a member of the mitogen-activated protein kinase family of kinases and shares a highly conserved kinase domain with other mitogen-activated protein kinase family members. The activation of NLK contributes to the pathogenesis of Diamond-Blackfan anemia (DBA), reducing c-myb expression and mechanistic target of rapamycin activity, and is therefore a potential therapeutic target. Unlike other anemias, the hematopoietic effects of DBA are largely restricted to the erythroid lineage. Mutations in ribosomal genes induce ribosomal insufficiency and reduced protein translation, dramatically impacting early erythropoiesis in the bone marrow of patients with DBA. We sought to identify compounds that suppress NLK and increases erythropoiesis in ribosomal insufficiency. We report that the active component of ginseng, ginsenoside Rb1, suppresses NLK expression and improves erythropoiesis in in vitro models of DBA. Ginsenoside Rb1-mediated suppression of NLK occurs through the upregulation of miR-208, which binds to the 3'-UTR of NLK mRNA and targets it for degradation. We also compare ginsenoside Rb1-mediated upregulation of miR-208 with metformin-mediated upregulation of miR-26. We conclude that targeting NLK expression through miRNA binding of the unique 3'-UTR is a viable alternative to the challenges of developing small-molecule inhibitors to target the highly conserved kinase domain of this specific kinase.


Subject(s)
Anemia, Diamond-Blackfan/pathology , Erythropoiesis/drug effects , Ginsenosides/pharmacology , Panax/chemistry , Protein Serine-Threonine Kinases/drug effects , 3' Untranslated Regions , Animals , Humans
18.
Am J Hematol ; 96(10): 1275-1286, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34310730

ABSTRACT

Hematopoietic cell transplantation (HCT) brings important alterations in erythropoiesis and iron metabolism. Hepcidin, which regulates iron metabolism, increases in iron overload or inflammation and decreases with iron deficiency or activated erythropoiesis. Erythroferrone (ERFE) is the erythroid regulator of hepcidin. We investigated erythropoiesis and iron metabolism after allogeneic HCT in 70 patients randomized between erythropoietin (EPO) treatment or no EPO, by serially measuring hepcidin, ERFE, CRP (inflammation), soluble transferrin receptor (sTfR, erythropoiesis), serum iron and transferrin saturation (Tsat; iron for erythropoiesis) and ferritin (iron stores). We identified biological and clinical factors associated with serum hepcidin and ERFE levels. Serum ERFE correlated overall with sTfR and reticulocytes and inversely with hepcidin. Erythroferrone paralleled sTfR levels, dropping during conditioning and recovering with engraftment. Inversely, hepcidin peaked after conditioning and decreased during engraftment. Erythroferrone and hepcidin were not significantly different with or without EPO. Multivariate analyses showed that the major determinant of ERFE was erythropoiesis (sTfR, reticulocytes or serum Epo). Pretransplant hepcidin was associated with previous RBC transfusions and ferritin. After transplantation, the major determinants of hepcidin were iron status (ferritin at all time points and Tsat at day 56) and erythropoiesis (sTfR or reticulocytes or ERFE), while the impact of inflammation was less clear and clinical parameters had no detectable influence. Hepcidin remained significantly higher in patients with high compared to low pretransplant ferritin. After allogeneic HCT with or without EPO therapy, significant alterations of hepcidin occur between pretransplant and day 180, in correlation with iron status and inversely with erythroid ERFE.


Subject(s)
Erythropoiesis , Hematopoietic Stem Cell Transplantation , Hepcidins/metabolism , Iron/metabolism , Peptide Hormones/metabolism , Adult , Aged , Erythropoiesis/drug effects , Erythropoietin/therapeutic use , Female , Hepcidins/blood , Humans , Iron/blood , Male , Middle Aged , Peptide Hormones/blood , Transplantation, Homologous
19.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281163

ABSTRACT

Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.


Subject(s)
Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/physiology , Animals , Cell Differentiation/drug effects , Epigenomics/methods , Erythropoiesis/drug effects , Erythropoietin/physiology , Humans , Janus Kinase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Erythropoietin/physiology , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcriptome/genetics
20.
Int J Mol Sci ; 22(13)2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34281283

ABSTRACT

ß-thalassaemia is a rare genetic condition caused by mutations in the ß-globin gene that result in severe iron-loading anaemia, maintained by a detrimental state of ineffective erythropoiesis (IE). The role of multiple mechanisms involved in the pathophysiology of the disease has been recently unravelled. The unbalanced production of α-globin is a major source of oxidative stress and membrane damage in red blood cells (RBC). In addition, IE is tightly linked to iron metabolism dysregulation, and the relevance of new players of this pathway, i.e., hepcidin, erythroferrone, matriptase-2, among others, has emerged. Advances have been made in understanding the balance between proliferation and maturation of erythroid precursors and the role of specific factors in this process, such as members of the TGF-ß superfamily, and their downstream effectors, or the transcription factor GATA1. The increasing understanding of IE allowed for the development of a broad set of potential therapeutic options beyond the current standard of care. Many candidates of disease-modifying drugs are currently under clinical investigation, targeting the regulation of iron metabolism, the production of foetal haemoglobin, the maturation process, or the energetic balance and membrane stability of RBC. Overall, they provide tools and evidence for multiple and synergistic approaches that are effectively moving clinical research in ß-thalassaemia from bench to bedside.


Subject(s)
Erythropoiesis/drug effects , Erythropoiesis/physiology , beta-Thalassemia/drug therapy , beta-Thalassemia/physiopathology , Activin Receptors, Type II/therapeutic use , Drug Development , GATA1 Transcription Factor/metabolism , Hepcidins/therapeutic use , Humans , Immunoglobulin Fc Fragments/therapeutic use , Iron/metabolism , Models, Biological , Mutation , Piperazines/therapeutic use , Quinolines/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor beta/metabolism , beta-Globins/genetics , beta-Thalassemia/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...