Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Cell ; 168(3): 344-361, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28129536

RESUMO

200 billion red blood cells (RBCs) are produced every day, requiring more than 2 × 1015 iron atoms every second to maintain adequate erythropoiesis. These numbers translate into 20 mL of blood being produced each day, containing 6 g of hemoglobin and 20 mg of iron. These impressive numbers illustrate why the making and breaking of RBCs is at the heart of iron physiology, providing an ideal context to discuss recent progress in understanding the systemic and cellular mechanisms that underlie the regulation of iron homeostasis and its disorders.


Assuntos
Células Eritroides/metabolismo , Ferro/metabolismo , Anemia/metabolismo , Animais , Transporte Biológico , Eritropoese , Hepcidinas/metabolismo , Humanos , Inflamação/metabolismo , Ferro da Dieta/metabolismo
2.
Cell ; 158(4): 849-860, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126789

RESUMO

Distal enhancers commonly contact target promoters via chromatin looping. In erythroid cells, the locus control region (LCR) contacts ß-type globin genes in a developmental stage-specific manner to stimulate transcription. Previously, we induced LCR-promoter looping by tethering the self-association domain (SA) of Ldb1 to the ß-globin promoter via artificial zinc fingers. Here, we show that targeting the SA to a developmentally silenced embryonic globin gene in adult murine erythroblasts triggers its transcriptional reactivation. This activity depends on the LCR, consistent with an LCR-promoter looping mechanism. Strikingly, targeting the SA to the fetal γ-globin promoter in primary adult human erythroblasts increases γ-globin promoter-LCR contacts, stimulating transcription to approximately 85% of total ß-globin synthesis, with a reciprocal reduction in adult ß-globin expression. Our findings demonstrate that forced chromatin looping can override a stringent developmental gene expression program and suggest a novel approach to control the balance of globin gene transcription for therapeutic applications.


Assuntos
Cromatina/metabolismo , Hemoglobina Fetal/genética , Técnicas Genéticas , Região de Controle de Locus Gênico , Ativação Transcricional , Globinas beta/genética , Animais , Antígenos CD34/metabolismo , Cromatina/química , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Humanos , Camundongos , Cultura Primária de Células
3.
Blood ; 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38949981

RESUMO

-Thalassemia (AT) is one of the most commonly occurring inherited hematological diseases. However, few treatments are available, and allogeneic bone marrow transplantation (BMT) is the only available therapeutic option for patients with severe AT. Research into AT has remained limited due to a lack of adult mouse models, with severe AT typically resulting in in utero lethality. By using a lipid nanoparticle (LNP) targeting the receptor CD117 and delivering a Cre mRNA (mRNACreLNPCD117), we were able to delete floxed -globin genes at high efficiency in hematopoietic stem cells (HSC) ex vivo. These cells were then engrafted in the absence or presence of a novel α-globin expressing lentiviral vector (ALS20I). Myeloablated mice transplanted with mRNACreLNPCD117-treated HSC showed a complete knockout of -globin genes. They demonstrated a phenotype characterized by the synthesis of hemoglobin H (-tetramers,  or HbH), aberrant erythropoiesis, and abnormal organ morphology, culminating in lethality approximately eight weeks following engraftment. Mice receiving mRNACreLNPCD117-treated HSC with at least one copy of ALS20I survived long-term with normalization of erythropoiesis, decreased the production of HbH, and ameliorated the abnormal organ morphology. Furthermore, we tested ALS20I in erythroid progenitors derived from -globin-KO CD34+ and cells isolated from patients with both deletional and non-deletional HbH disease, demonstrating improvement in -globin/-globin mRNA ratio and reduction in the formation of HbH by HPLC. Our results demonstrate the broad applicability of LNP for disease modeling, characterization of a novel severe mouse model of AT, and the efficacy of ALS20I for treating AT.

4.
Am J Hematol ; 99(7): 1300-1312, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38659383

RESUMO

A recently approved drug that induces erythroid cell maturation (luspatercept) has been shown to improve anemia and reduce the need for blood transfusion in non-transfusion-dependent as well as transfusion-dependent ß-thalassemia (BT) patients. Although these results were predominantly positive, not all the patients showed the expected increase in hemoglobin (Hb) levels or transfusion burden reduction. Additional studies indicated that administration of luspatercept in transfusion-dependent BT was associated with increased erythropoietic markers, decreased hepcidin levels, and increased liver iron content. Altogether, these studies suggest that luspatercept may necessitate additional drugs for improved erythroid and iron management. As luspatercept does not appear to directly affect iron metabolism, we hypothesized that TMPRSS6-ASO could improve iron parameters and iron overload when co-administered with luspatercept. We used an agent analogous to murine luspatercept (RAP-GRL) and another novel therapeutic, IONIS TMPRSS6-LRx (TMPRSS6-ASO), a hepcidin inducer, to treat non-transfusion-dependent BT-intermedia mice. Our study shows that RAP-GRL alone improved red blood cell (RBC) production, with no or limited effect on splenomegaly and iron parameters. In contrast, TMPRSS6-ASO improved RBC measurements, ameliorated splenomegaly, and improved iron overload most effectively. Our results provide pre-clinical support for combining TMPRSS6-ASO and luspatercept in treating BT, as these drugs together show potential for simultaneously improving both erythroid and iron parameters in BT patients.


Assuntos
Proteínas de Membrana , Serina Endopeptidases , Talassemia beta , Talassemia beta/tratamento farmacológico , Talassemia beta/terapia , Camundongos , Animais , Proteínas de Membrana/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Fragmentos Fc das Imunoglobulinas/farmacologia , Humanos , Fator de Crescimento Transformador beta/metabolismo , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/etiologia , Hepcidinas , Ferro/metabolismo , Feminino , Masculino , Quimioterapia Combinada , Receptores de Activinas Tipo II
5.
Am J Hematol ; 99(7): 1349-1359, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38400590

RESUMO

Primum non nocere! Can iron deficiency, an abnormality that causes anemia, benefit people with sickle cell disease (SCD) who already have an anemia? The published literature we review appears to answer this question in the affirmative: basic science considerations, animal model experiments, and noncontrolled clinical observations all suggest a therapeutic potential of iron restriction in SCD. This is because SCD's clinical manifestations are ultimately attributable to the polymerization of hemoglobin S (HbS), a process strongly influenced by intracellular HbS concentration. Even small decrements in HbS concentration greatly reduce polymerization, and iron deficiency lowers erythrocyte hemoglobin concentration. Thus, iron deficiency could improve SCD by changing its clinical features to those of a more benign anemia (i.e., a condition with fewer or no vaso-occlusive events). We propose that well-designed clinical studies be implemented to definitively determine whether iron restriction is a safe and effective option in SCD. These investigations are particularly timely now that pharmacologic agents are being developed, which may directly reduce red cell hemoglobin concentrations without the need for phlebotomies to deplete total body iron.


Assuntos
Anemia Falciforme , Hemoglobina Falciforme , Ferro , Anemia Falciforme/complicações , Anemia Falciforme/sangue , Humanos , Animais , Ferro/metabolismo , Ferro/sangue , Hemoglobina Falciforme/metabolismo , Hemoglobina Falciforme/análise , Anemia Ferropriva/tratamento farmacológico , Eritrócitos/metabolismo
6.
Haematologica ; 108(10): 2582-2593, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37345473

RESUMO

Under normal conditions, iron metabolism is carefully regulated to sustain normal cellular functions and the production of hemoglobin in erythroid cells. Perturbation to the erythropoiesis-iron metabolism axis can result in iron imbalances and cause anemia or organ toxicity. Various congenital and acquired diseases associated with abnormal red cell production are characterized by aberrant iron absorption. Several recent studies have shown that improvements in red blood cell production also ameliorate iron metabolism and vice versa. Many therapeutics are now under development with the potential to improve a variety of hematologic diseases, from ß-thalassemia and iron-refractory iron deficiency anemia to anemia of inflammation and polycythemia vera. This review summarizes selected mechanisms related to red cell production and iron metabolism and describes potential therapeutics and their current uses. We also consider the potential application of the discussed therapeutics on various diseases, alone or in combination. The vast repertoire of drugs under development offers new opportunities to improve the clinical care of patients suffering from congenital or acquired red blood cell disorders with limited or no treatment options.


Assuntos
Anemia Ferropriva , Doenças Hematológicas , Talassemia beta , Humanos , Eritropoese , Eritrócitos/metabolismo , Ferro/metabolismo , Talassemia beta/metabolismo , Doenças Hematológicas/tratamento farmacológico
7.
Blood ; 136(17): 1968-1979, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32556142

RESUMO

ß-Thalassemia intermedia is a disorder characterized by ineffective erythropoiesis (IE), anemia, splenomegaly, and systemic iron overload. Novel approaches are being explored based on the modulation of pathways that reduce iron absorption (ie, using hepcidin activators like Tmprss6-antisense oligonucleotides [ASOs]) or increase erythropoiesis (by erythropoietin [EPO] administration or modulating the ability of transferrin receptor 2 [Tfr2] to control red blood cell [RBC] synthesis). Targeting Tmprss6 messenger RNA by Tmprss6-ASO was proven to be effective in improving IE and splenomegaly by inducing iron restriction. However, we postulated that combinatorial strategies might be superior to single therapies. Here, we combined Tmprss6-ASO with EPO administration or removal of a single Tfr2 allele in the bone marrow of animals affected by ß-thalassemia intermedia (Hbbth3/+). EPO administration alone or removal of a single Tfr2 allele increased hemoglobin levels and RBCs. However, EPO or Tfr2 single-allele deletion alone, respectively, exacerbated or did not improve splenomegaly in ß-thalassemic mice. To overcome this issue, we postulated that some level of iron restriction (by targeting Tmprss6) would improve splenomegaly while preserving the beneficial effects on RBC production mediated by EPO or Tfr2 deletion. While administration of Tmprss6-ASO alone improved the anemia, the combination of Tmprss6-ASO + EPO or Tmprss6-ASO + Tfr2 single-allele deletion produced significantly higher hemoglobin levels and reduced splenomegaly. In conclusion, our results clearly indicate that these combinatorial approaches are superior to single treatments in ameliorating IE and anemia in ß-thalassemia and could provide guidance to translate some of these approaches into viable therapies.


Assuntos
Eritropoetina/administração & dosagem , Eritropoetina/genética , Terapia Genética/métodos , Proteínas de Membrana/antagonistas & inibidores , Oligonucleotídeos Antissenso/administração & dosagem , Talassemia beta/terapia , Animais , Células Cultivadas , Eritropoese/efeitos dos fármacos , Eritropoese/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Ferro/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/prevenção & controle , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligonucleotídeos Antissenso/farmacologia , Receptores da Transferrina/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Talassemia beta/metabolismo
8.
Am J Hematol ; 97(10): 1324-1336, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36071579

RESUMO

ß-thalassemia is a genetic disorder caused by mutations in the ß-globin gene, and characterized by anemia, ineffective erythropoiesis and iron overload. Patients affected by the most severe transfusion-dependent form of the disease (TDT) require lifelong blood transfusions and iron chelation therapy, a symptomatic treatment associated with several complications. Other therapeutic opportunities are available, but none is fully effective and/or applicable to all patients, calling for the identification of novel strategies. Transferrin receptor 2 (TFR2) balances red blood cells production according to iron availability, being an activator of the iron-regulatory hormone hepcidin in the liver and a modulator of erythropoietin signaling in erythroid cells. Selective Tfr2 deletion in the BM improves anemia and iron-overload in non-TDT mice, both as a monotherapy and, even more strikingly, in combination with iron-restricting approaches. However, whether Tfr2 targeting might represent a therapeutic option for TDT has never been investigated so far. Here, we prove that BM Tfr2 deletion improves anemia, erythrocytes morphology and ineffective erythropoiesis in the Hbbth1/th2 murine model of TDT. This effect is associated with a decrease in the expression of α-globin, which partially corrects the unbalance with ß-globin chains and limits the precipitation of misfolded hemoglobin, and with a decrease in the activation of unfolded protein response. Remarkably, BM Tfr2 deletion is also sufficient to avoid long-term blood transfusions required for survival of Hbbth1/th2 animals, preventing mortality due to chronic anemia and reducing transfusion-associated complications, such as progressive iron-loading. Altogether, TFR2 targeting might represent a promising therapeutic option also for TDT.


Assuntos
Sobrecarga de Ferro , Receptores da Transferrina , Talassemia beta , Animais , Transfusão de Sangue , Modelos Animais de Doenças , Ferro/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Camundongos , Receptores da Transferrina/genética , Globinas beta , Talassemia beta/genética , Talassemia beta/terapia
9.
Mol Ther ; 29(4): 1625-1638, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33515514

RESUMO

Ongoing clinical trials for treatment of beta-globinopathies by gene therapy involve the transfer of the beta-globin gene, which requires integration of three to four copies per genome in most target cells. This high proviral load may increase genome toxicity, potentially limiting the safety of this therapy and relegating its use to total body myeloablation. We hypothesized that introducing an additional hypersensitive site from the locus control region, the complete sequence of the second intron of the beta-globin gene, and the ankyrin insulator may enhance beta-globin expression. We identified a construct, ALS20, that synthesized significantly higher adult hemoglobin levels than those of other constructs currently used in clinical trials. These findings were confirmed in erythroblastic cell lines and in primary cells isolated from sickle cell disease patients. Bone marrow transplantation studies in beta-thalassemia mice revealed that ALS20 was curative at less than one copy per genome. Injection of human CD34+ cells transduced with ALS20 led to safe, long-term, and high polyclonal engraftment in xenograft experiments. Successful treatment of beta-globinopathies with ALS20 could potentially be achieved at less than two copies per genome, minimizing the risk of cytotoxic events and lowering the intensity of myeloablation.


Assuntos
Anemia Falciforme/genética , Transplante de Medula Óssea , Terapia Genética , Globinas beta/genética , Talassemia beta/genética , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Animais , Expressão Gênica/genética , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Hemoglobinas/genética , Xenoenxertos , Humanos , Lentivirus/genética , Região de Controle de Locus Gênico/genética , Camundongos , Transdução Genética , Globinas beta/uso terapêutico , Talassemia beta/sangue , Talassemia beta/patologia , Talassemia beta/terapia
10.
Hum Mol Genet ; 28(R1): R24-R30, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31322165

RESUMO

Recently, gene therapy clinical trials have been successfully applied to hemoglobinopathies, such as sickle cell disease (SCD) and ß-thalassemia. Among the great discoveries that led to the design of genetic approaches to cure these disorders is the discovery of the ß-globin locus control region and several associated transcription factors, which determine hemoglobin switching as well as high-level, erythroid-specific expression of genes at the ß-globin locus. Moreover, increasing evidence shows that lentiviral vectors are efficient tools to insert large DNA elements into nondividing hematopoietic stem cells, showing reassuring safe integration profiles. Alternatively, genome editing could restore expression of fetal hemoglobin or target specific mutations to restore expression of the wild-type ß-globin gene. The most recent clinical trials for ß-thalassemia and SCD are showing promising outcomes: patients were able to discontinue transfusions or had reduced transfusion requirements. However, toxic myeloablation and the high cost of current ex vivo hematopoietic stem cell gene therapy platforms represent a barrier to a widespread application of these approaches. In this review, we summarize these gene therapy strategies and ongoing clinical trials. Finally, we discuss possible strategies to improve outcomes, reduce myeloablative regimens and future challenges to reduce the cost of gene therapy platform.


Assuntos
Terapia Genética , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Animais , Ensaios Clínicos como Assunto , Regulação da Expressão Gênica , Predisposição Genética para Doença , Terapia Genética/efeitos adversos , Terapia Genética/economia , Terapia Genética/métodos , Terapia Genética/tendências , Vetores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas , Hemoglobinas/genética , Humanos , Mutação , Transdução Genética , Resultado do Tratamento
11.
Am J Pathol ; 190(10): 2146-2154, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32745462

RESUMO

Patients with thalassemia exhibit an increased risk of thrombotic events that is augmented after splenectomy. Heparanase protein enhances cancer progression, angiogenesis, and inflammation; it also activates the coagulation system through direct interaction with tissue factor (TF). Additionally, erythropoietin, which is elevated in anemic patients, up-regulates heparanase expression via the Janus kinase 2 (JAK-2) pathway. This study aimed was to explore the heparanase profile in thalassemia. Coagulation factors were analyzed via immunostaining, enzyme-linked immunosorbent assay, and heparanase procoagulant activity assay. In spleen specimens of thalassemia major patients, a higher level of heparanase staining was observed compared with control spleens resected after trauma (P < 0.001). Higher heparanase levels, heparanase and TF procoagulant activity, and erythropoietin levels were found in the plasma of 67 thalassemia major patients compared with 29 control subjects. No difference was found in pediatric patients (23 of 67) compared with adults or splenectomized versus nonsplenectomized patients. Higher levels of heparanase, TF, TF pathway inhibitor, and TF pathway inhibitor-2 were observed in liver, spleen, heart, and kidney tissues of thalassemia intermedia mice (Hbbth3/+). These protein levels significantly reduced when mice were treated with the JAK-2 inhibitor ruxolitinib (P < 0.0001). In summary, heparanase levels are elevated in thalassemia, which may contribute to thrombotic phenomena in these patients. Inhibition of heparanase or the JAK-2 pathway may reduce thrombotic risk in thalassemia.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Glucuronidase/metabolismo , Janus Quinase 2/antagonistas & inibidores , Lipoproteínas/farmacologia , Trombose/tratamento farmacológico , Adulto , Animais , Humanos , Masculino , Camundongos Endogâmicos C57BL , Talassemia/tratamento farmacológico , Talassemia/metabolismo , Tromboplastina/metabolismo , Trombose/metabolismo , Adulto Jovem
12.
Blood ; 133(1): 51-58, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30401707

RESUMO

ß-Thalassemia (BT) is an inherited genetic disorder that is characterized by ineffective erythropoiesis (IE), leading to anemia and abnormal iron metabolism. IE is an abnormal expansion of the number of erythroid progenitor cells with unproductive synthesis of enucleated erythrocytes, leading to anemia and hypoxia. Anemic patients affected by BT suffer from iron overload, even in the absence of chronic blood transfusion, suggesting the presence of ≥1 erythroid factor with the ability to modulate iron metabolism and dietary iron absorption. Recent studies suggest that decreased erythroid cell differentiation and survival also contribute to IE, aggravating the anemia in BT. Furthermore, hypoxia can also affect and increase iron absorption. Understanding the relationship between iron metabolism and IE could provide important insights into the BT condition and help to develop novel treatments. In fact, genetic or pharmacological manipulations of iron metabolism or erythroid cell differentiation and survival have been shown to improve IE, iron overload, and anemia in animal models of BT. Based on those findings, new therapeutic approaches and drugs have been proposed; clinical trials are underway that have the potential to improve erythrocyte production, as well as to reduce the iron overload and organ toxicity in BT and in other disorders characterized by IE.


Assuntos
Eritropoese , Distúrbios do Metabolismo do Ferro/etiologia , Distúrbios do Metabolismo do Ferro/patologia , Ferro/metabolismo , Talassemia beta/complicações , Animais , Humanos , Talassemia beta/patologia
13.
Blood ; 134(17): 1373-1384, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31434707

RESUMO

Transferrin, the major plasma iron-binding molecule, interacts with cell-surface receptors to deliver iron, modulates hepcidin expression, and regulates erythropoiesis. Transferrin binds and releases iron via either or both of 2 homologous lobes (N and C). To test the hypothesis that the specificity of iron occupancy in the N vs C lobe influences transferrin function, we generated mice with mutations to abrogate iron binding in either lobe (TfN-bl or TfC-bl). Mice homozygous for either mutation had hepatocellular iron loading and decreased liver hepcidin expression (relative to iron concentration), although to different magnitudes. Both mouse models demonstrated some aspects of iron-restricted erythropoiesis, including increased zinc protoporphyrin levels, decreased hemoglobin levels, and microcytosis. Moreover, the TfN-bl/N-bl mice demonstrated the anticipated effect of iron restriction on red cell production (ie, no increase in red blood cell [RBC] count despite elevated erythropoietin levels), along with a poor response to exogenous erythropoietin. In contrast, the TfC-bl/C-bl mice had elevated RBC counts and an exaggerated response to exogenous erythropoietin sufficient to ameliorate the anemia. Observations in heterozygous mice further support a role for relative N vs C lobe iron occupancy in transferrin-mediated regulation of iron homeostasis and erythropoiesis.


Assuntos
Eritropoese , Ferro/metabolismo , Transferrina/metabolismo , Animais , Sítios de Ligação , Contagem de Eritrócitos , Eritropoetina/metabolismo , Feminino , Homeostase , Masculino , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transferrina/química , Transferrina/genética
14.
Int J Mol Sci ; 22(3)2021 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498614

RESUMO

Oxidative damage by reactive oxygen species (ROS) is one of the main contributors to cell injury and tissue damage in thalassemia patients. Recent studies suggest that ROS generation in non-transfusion-dependent (NTDT) patients occurs as a result of iron overload. Among the different sources of ROS, the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes and cytochrome P450 (CYP450) have been proposed to be major contributors for oxidative stress in several diseases. However, the sources of ROS in patients with NTDT remain poorly understood. In this study, Hbbth3/+ mice, a mouse model for ß-thalassemia, were used. These mice exhibit an unchanged or decreased expression of the major NOX isoforms, NOX1, NOX2 and NOX4, when compared to their C57BL/6 control littermates. However, a significant increase in the protein synthesis of CYP4A and CYP4F was observed in the Hbbth3/+ mice when compared to the C57BL/6 control mice. These changes were paralleled by an increased production of 20-hydroxyeicosatetraenoic acid (20-HETE), a CYP4A and CYP4F metabolite. Furthermore, these changes corroborate with onset of ROS production concomitant with liver injury. To our knowledge, this is the first report indicating that CYP450 4A and 4F-induced 20-HETE production mediates reactive oxygen species overgeneration in Hbbth3/+ mice through an NADPH-dependent pathway.


Assuntos
Família 4 do Citocromo P450/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Talassemia beta/metabolismo , Animais , Modelos Animais de Doenças , Hepatite/etiologia , Hepatite/patologia , Ferro/metabolismo , Isoenzimas/metabolismo , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/metabolismo , Talassemia beta/complicações , Talassemia beta/patologia
15.
Kidney Int ; 98(5): 1210-1224, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32574618

RESUMO

Anemia is a frequent complication of chronic kidney disease (CKD), related in part to the disruption of iron metabolism. Iron therapy is very common in children with CKD and excess iron has been shown to induce bone loss in non-CKD settings, but the impact of iron on bone health in CKD remains poorly understood. Here, we evaluated the effect of oral and parenteral iron therapy on bone transcriptome, bone histology and morphometry in two mouse models of juvenile CKD (adenine-induced and 5/6-nephrectomy). Both modalities of iron therapy effectively improved anemia in the mice with CKD, and lowered bone Fgf23 expression. At the same time, iron therapy suppressed genes implicated in bone formation and resulted in the loss of cortical and trabecular bone in the mice with CKD. Bone resorption was activated in untreated CKD, but iron therapy had no additional effect on this. Furthermore, we assessed the relationship between biomarkers of bone turnover and iron status in a cohort of children with CKD. Children treated with iron had lower levels of circulating biomarkers of bone formation (bone-specific alkaline phosphatase and the amino-terminal propeptide of type 1 procollagen), as well as fewer circulating osteoblast precursors, compared to children not treated with iron. These differences were independent of age, sex, and glomerular filtration rate. Thus, iron therapy adversely affected bone health in juvenile mice with CKD and was associated with low levels of bone formation biomarkers in children with CKD.


Assuntos
Dextranos , Insuficiência Renal Crônica , Animais , Densidade Óssea , Fator de Crescimento de Fibroblastos 23 , Taxa de Filtração Glomerular , Ferro , Camundongos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico
16.
Blood ; 131(16): 1790-1794, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29523504

RESUMO

Hepcidin agonists are a new class of compounds that regulate blood iron levels, limit iron absorption, and could improve the treatment of hemochromatosis, ß-thalassemia, polycythemia vera, and other disorders in which disrupted iron homeostasis causes or contributes to disease. Hepcidin agonists also have the potential to prevent severe complications of siderophilic infections in patients with iron overload or chronic liver disease. This review highlights the preclinical studies that support the development of hepcidin agonists for the treatment of these disorders.


Assuntos
Hemocromatose/tratamento farmacológico , Hepcidinas/agonistas , Policitemia Vera/tratamento farmacológico , Talassemia beta/tratamento farmacológico , Animais , Hemocromatose/metabolismo , Hemocromatose/patologia , Hepcidinas/metabolismo , Humanos , Ferro/metabolismo , Policitemia Vera/metabolismo , Policitemia Vera/patologia , Talassemia beta/metabolismo , Talassemia beta/patologia
17.
Blood ; 131(8): 899-910, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29237594

RESUMO

Nonclassical ferroportin disease (FD) is a form of hereditary hemochromatosis caused by mutations in the iron transporter ferroportin (Fpn), resulting in parenchymal iron overload. Fpn is regulated by the hormone hepcidin, which induces Fpn endocytosis and cellular iron retention. We characterized 11 clinically relevant and 5 nonclinical Fpn mutations using stably transfected, inducible isogenic cell lines. All clinical mutants were functionally resistant to hepcidin as a consequence of either impaired hepcidin binding or impaired hepcidin-dependent ubiquitination despite intact hepcidin binding. Mapping the residues onto 2 computational models of the human Fpn structure indicated that (1) mutations that caused ubiquitination-resistance were positioned at helix-helix interfaces, likely preventing the hepcidin-induced conformational change, (2) hepcidin binding occurred within the central cavity of Fpn, (3) hepcidin interacted with up to 4 helices, and (4) hepcidin binding should occlude Fpn and interfere with iron export independently of endocytosis. We experimentally confirmed hepcidin-mediated occlusion of Fpn in the absence of endocytosis in multiple cellular systems: HEK293 cells expressing an endocytosis-defective Fpn mutant (K8R), Xenopus oocytes expressing wild-type or K8R Fpn, and mature human red blood cells. We conclude that nonclassical FD is caused by Fpn mutations that decrease hepcidin binding or hinder conformational changes required for ubiquitination and endocytosis of Fpn. The newly documented ability of hepcidin and its agonists to occlude iron transport may facilitate the development of broadly effective treatments for hereditary iron overload disorders.


Assuntos
Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Resistência a Medicamentos , Hepcidinas/metabolismo , Ferro/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Simulação por Computador , Endocitose , Células HEK293 , Hepcidinas/agonistas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Mutação , Oócitos/citologia , Oócitos/metabolismo , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Relação Estrutura-Atividade , Ubiquitinação , Xenopus laevis
18.
Haematologica ; 105(7): 1835-1844, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31582543

RESUMO

Minihepcidins are hepcidin agonists that have been previously shown to reverse iron overload and improve erythropoiesis in mice affected by non-transfusion-dependent thalassemia. Given the extreme anemia that occurred with the previous model of transfusion-dependent thalassemia, that model was inadequate for investigating whether minihepcidins can improve red blood cell quality, lifespan and ineffective erythropoiesis. To overcome this limitation, we generated a new murine model of transfusion-dependent thalassemia with severe anemia and splenomegaly, but sufficient red cells and hemoglobin production to test the effect of minihepcidins. Furthermore, this new model demonstrates cardiac iron overload for the first time. In the absence of transfusions, minihepcidins improved red blood cell morphology and lifespan as well as ineffective erythropoiesis. Administration of a minihepcidin in combination with chronic red blood cell transfusion further improved the ineffective erythropoiesis and splenomegaly and reversed cardiac iron overload. These studies indicate that drugs such as minihepcidins have therapeutic potential for patients with transfusion-dependent thalassemia.


Assuntos
Hepcidinas/uso terapêutico , Sobrecarga de Ferro , Esplenomegalia , Talassemia beta , Animais , Modelos Animais de Doenças , Eritropoese , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/etiologia , Camundongos , Esplenomegalia/tratamento farmacológico , Esplenomegalia/etiologia , Talassemia beta/terapia
19.
Blood ; 130(9): 1144-1155, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28729432

RESUMO

Biogenesis of mammalian red blood cells requires nuclear expulsion by orthochromatic erythoblasts late in terminal differentiation (enucleation), but the mechanism is largely unexplained. Here, we employed high-resolution confocal microscopy to analyze nuclear morphology and F-actin rearrangements during the initiation, progression, and completion of mouse and human erythroblast enucleation in vivo. Mouse erythroblast nuclei acquire a dumbbell-shaped morphology during enucleation, whereas human bone marrow erythroblast nuclei unexpectedly retain their spherical morphology. These morphological differences are linked to differential expression of Lamin isoforms, with primary mouse erythroblasts expressing only Lamin B and primary human erythroblasts only Lamin A/C. We did not consistently identify a continuous F-actin ring at the cell surface constriction in mouse erythroblasts, nor at the membrane protein-sorting boundary in human erythroblasts, which do not have a constriction, arguing against a contractile ring-based nuclear expulsion mechanism. However, both mouse and human erythroblasts contain an F-actin structure at the rear of the translocating nucleus, enriched in tropomodulin 1 (Tmod1) and nonmuscle myosin IIB. We investigated Tmod1 function in mouse and human erythroblasts both in vivo and in vitro and found that absence of Tmod1 leads to enucleation defects in mouse fetal liver erythroblasts, and in CD34+ hematopoietic stem and progenitor cells, with increased F-actin in the structure at the rear of the nucleus. This novel structure, the "enucleosome," may mediate common cytoskeletal mechanisms underlying erythroblast enucleation, notwithstanding the morphological heterogeneity of enucleation across species.


Assuntos
Actinas/metabolismo , Núcleo Celular/metabolismo , Eritroblastos/metabolismo , Tropomodulina/metabolismo , Animais , Medula Óssea/metabolismo , Diferenciação Celular , Forma do Núcleo Celular , Polaridade Celular , Feto/metabolismo , Técnicas de Silenciamento de Genes , Laminas/metabolismo , Fígado/embriologia , Camundongos Endogâmicos C57BL , Miosina não Muscular Tipo IIB/metabolismo , Isoformas de Proteínas/metabolismo
20.
Blood ; 129(11): 1514-1526, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28151426

RESUMO

Iron availability for erythropoiesis and its dysregulation in ß-thalassemia are incompletely understood. We previously demonstrated that exogenous apotransferrin leads to more effective erythropoiesis, decreasing erythroferrone (ERFE) and derepressing hepcidin in ß-thalassemic mice. Transferrin-bound iron binding to transferrin receptor 1 (TfR1) is essential for cellular iron delivery during erythropoiesis. We hypothesize that apotransferrin's effect is mediated via decreased TfR1 expression and evaluate TfR1 expression in ß-thalassemic mice in vivo and in vitro with and without added apotransferrin. Our findings demonstrate that ß-thalassemic erythroid precursors overexpress TfR1, an effect that can be reversed by the administration of exogenous apotransferrin. In vitro experiments demonstrate that apotransferrin inhibits TfR1 expression independent of erythropoietin- and iron-related signaling, decreases TfR1 partitioning to reticulocytes during enucleation, and enhances enucleation of defective ß-thalassemic erythroid precursors. These findings strongly suggest that overexpressed TfR1 may play a regulatory role contributing to iron overload and anemia in ß-thalassemic mice. To evaluate further, we crossed TfR1+/- mice, themselves exhibiting iron-restricted erythropoiesis with increased hepcidin, with ß-thalassemic mice. Resultant double-heterozygote mice demonstrate long-term improvement in ineffective erythropoiesis, hepcidin derepression, and increased erythroid enucleation in relation to ß-thalassemic mice. Our data demonstrate for the first time that TfR1+/- haploinsufficiency reverses iron overload specifically in ß-thalassemic erythroid precursors. Taken together, decreasing TfR1 expression during ß-thalassemic erythropoiesis, either directly via induced haploinsufficiency or via exogenous apotransferrin, decreases ineffective erythropoiesis and provides an endogenous mechanism to upregulate hepcidin, leading to sustained iron-restricted erythropoiesis and preventing systemic iron overload in ß-thalassemic mice.


Assuntos
Anemia/etiologia , Hepcidinas/metabolismo , Receptores da Transferrina/metabolismo , Talassemia beta/metabolismo , Anemia/prevenção & controle , Animais , Apoproteínas/administração & dosagem , Apoproteínas/farmacocinética , Eritropoese , Sobrecarga de Ferro/etiologia , Camundongos , Transferrina/administração & dosagem , Transferrina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA