Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
NMR Biomed ; 30(5)2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28186661

RESUMO

To characterize the cerebral profile associated with sickle cell disease (SCD), we used in vivo proton MRI and MRS to quantify hemodynamics and neurochemicals in the thalamus of NY1DD mice, a mild model of SCD, and compared them with wild-type (WT) control mice. Compared with WT mice, NY1DD mice at steady state had elevated cerebral blood flow (CBF) and concentrations of N-acetylaspartate (NAA), glutamate (Glu), alanine, total creatine and N-acetylaspartylglutamate. Concentrations of glutathione (GSH) at steady state showed a negative correlation with BOLD signal change in response to 100% oxygen, a marker for oxidative stress, and mean diffusivity assessed using diffusion-tensor imaging, a marker for edematous inflammation. In NY1DD mice, elevated basal CBF was correlated negatively with [NAA], but positively with concentration of glutamine ([Gln]). Immediately after experimental hypoxia (at reoxygenation after 18 hours of 8% O2 ), concentrations of NAA, Glu, GSH, Gln and taurine (Tau) increased only in NY1DD mice. [NAA], [Glu], [GSH] and [Tau] all returned to baseline levels two weeks after the hypoxic episode. The altered neurochemical profile in the NY1DD mouse model of SCD at steady state and following experimental hypoxia/reoxygenation suggests a state of chronic oxidative stress leading to compensatory cerebral metabolic adjustments.


Assuntos
Anemia Falciforme/fisiopatologia , Biopolímeros/metabolismo , Velocidade do Fluxo Sanguíneo , Encéfalo/fisiopatologia , Circulação Cerebrovascular , Imageamento por Ressonância Magnética , Espectroscopia de Prótons por Ressonância Magnética , Anemia Falciforme/diagnóstico , Animais , Biomarcadores/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Imagem Molecular , Consumo de Oxigênio
2.
Am J Physiol Heart Circ Physiol ; 304(1): H42-50, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23125209

RESUMO

Chronic inflammation is a salient feature of sickle cell disease (SCD) and transgenic-knockout sickle (BERK) mice. Inflammation is implicated in the activation of hypoxia-inducible factor-1α (HIF-1α) under normoxic conditions. We hypothesize that, in SCD, inflammation coupled with nitric oxide (NO) depletion will induce expression of HIF-1α, a transcription factor with wide-ranging effects including activation of genes for vasoactive molecules. To this end, we have examined the expression of HIF-1α in normoxic BERK mice expressing exclusively human α- and ß(S)- globins, and evaluated the effect of fetal hemoglobin (HbF) in BERK mice (i.e., <1.0%, 20%, and 40% HbF). HbF exerts antisickling and anti-inflammatory effects. Here, we show that HIF-1α is expressed in BERK mice under normoxic conditions, accompanied by increased expression of its vasoactive biomarkers such as VEGF, heme oxygenase-1 (HO-1), and serum ET-1 levels. In BERK mice expressing HbF, HIF-1α expression decreases concomitantly with increasing HbF, commensurately with increased NO bioavailability, and shows a strong inverse correlation with plasma NO metabolites (NOx) levels. Reduced HIF-1α expression is associated with decreased HO-1, VEGF, and ET-1. Notably, arteriolar dilation, enhanced volumetric blood flow, and low blood pressure in normoxic BERK mice all show a trend toward normalization with the introduction of HbF. Also, arginine treatment reduced HIF-1α, as well as VEGF expression in normoxic BERK mice, supporting a role of NO bioavailability in HIF-1α activation. Thus HIF-1α expression in normoxic sickle mice is likely a consequence of chronic inflammation, and HbF exerts an ameliorating effect by decreasing sickling, increasing NO bioavailability, and reducing inflammation.


Assuntos
Anemia Falciforme/terapia , Arteríolas/metabolismo , Hemoglobina Fetal/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/terapia , Microcirculação , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/fisiopatologia , Animais , Arginina/farmacologia , Arteríolas/efeitos dos fármacos , Arteríolas/fisiopatologia , Pressão Sanguínea , Modelos Animais de Doenças , Regulação para Baixo , Endotelina-1/sangue , Hemoglobina Fetal/genética , Heme Oxigenase-1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/fisiopatologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microcirculação/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fluxo Sanguíneo Regional , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vasodilatação , alfa-Globinas/genética , alfa-Globinas/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
3.
Blood Cells Mol Dis ; 48(2): 91-101, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22260787

RESUMO

Hemoglobin (Hb) E (ß26 Glu→Lys) is the most common abnormal hemoglobin (Hb) variant in the world. Homozygotes for HbE are mildly thalassemic as a result of the alternate splice mutation and present with a benign clinical picture (microcytic and mildly anemic) with rare clinical symptoms. Given that the human red blood cell (RBC) contains both HbE and excess α-chains along with minor hemoglobins, the consequence of HbE alone on RBC pathophysiology has not been elucidated. This becomes critical for the highly morbid ß(E)-thalassemia disease. We have generated transgenic mice exclusively expressing human HbE (HbEKO) that exhibit the known aberrant splicing of ß(E) globin mRNA, but are essentially non-thalassemic as demonstrated by RBC α/ß (human) globin chain synthesis. These mice exhibit hematological characteristics similar to presentations in human EE individuals: microcytic RBC with low MCV and MCH but normal MCHC; target RBC; mild anemia with low Hb, HCT and mildly elevated reticulocyte levels and decreased osmotic fragility, indicating altered RBC surface area to volume ratio. These alterations are correlated with a mild RBC oxidative stress indicated by enhanced membrane lipid peroxidation, elevated zinc protoporphyrin levels, and by small but significant changes in cardiac function. The C57 (background) mouse and full KO mouse models expressing HbE with the presence of HbS or HbA are used as controls. In select cases, the HbA full KO mouse model is compared but found to be limited due to its RBC thalassemic characteristics. Since the HbEKO mouse RBC lacks an abundance of excess α-chains that would approximate a mouse thalassemia (or a human thalassemia), the results indicate that the observed in vivo RBC mild oxidative stress arises, at least in part, from the molecular consequences of the HbE mutation.


Assuntos
Hemoglobina E/genética , Hemoglobina E/metabolismo , Camundongos Transgênicos , Estresse Oxidativo , Animais , Cruzamento , Índices de Eritrócitos , Eritrócitos/metabolismo , Feminino , Hemoglobinas Anormais/genética , Hemoglobinas Anormais/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragilidade Osmótica , alfa-Globinas/biossíntese , Globinas beta/biossíntese
4.
Transl Res ; 244: 75-87, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35091127

RESUMO

Fetal hemoglobin (HbF) is known to lessen the severity of sickle cell disease (SCD), through reductions in peripheral vaso-occlusive disease and reduced risk for cerebrovascular events. However, the influence of HbF on oxygen delivery to high metabolism tissues like the brain, or its influence on cerebral perfusion, metabolism, inflammation or function have not been widely studied. We employed a Berkley mouse model (BERK) of SCD with gamma transgenes q3 expressing exclusively human α- and ßS-globins with varying levels of γ globin expression to investigate the effect of HbF expression on the brain using magnetic resonance imaging (MRI), MRI diffusion tensor imaging (DTI) and spectroscopy (MRS) and hematological parameters. Hematological parameters improved with increasing γ level expression, as did markers for brain metabolism, perfusion and inflammation. Brain microstructure assessed by DTI fractional anisotropy improved, while myo-inositol levels increased, suggesting improved microstructural integrity and reduced cell loss. Our results suggest that increasing γ levels not only improves sickle peripheral disease, but also improves brain perfusion and oxygen delivery while reducing brain inflammation while protecting brain microstructural integrity.


Assuntos
Anemia Falciforme , Hemoglobina Fetal , Anemia Falciforme/complicações , Animais , Circulação Cerebrovascular , Imagem de Tensor de Difusão , Hemoglobina Fetal/metabolismo , Hemoglobina Falciforme , Inflamação , Camundongos , Oxigênio
5.
Exp Hematol ; 75: 31-52.e15, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31176681

RESUMO

Many methods have been developed to produce cultured red blood cells (cRBCs) in vitro but translational applications have been hampered by high costs of production and by low rates of enucleation. We have developed R6 and IMIT, two chemically defined culture media and combined them into robust erythroid differentiation (RED) protocols to differentiate induced-pluripotent stem cells (iPSCs) and peripheral blood mononuclear cells (MNCs) into enucleated erythroid cells. The RED protocols do not require any albumin or animal components and require ten- to twentyfold less transferrin (Tf) than previously, because iron is provided to the differentiating erythroblasts by small amounts of recombinant Tf supplemented with FeIII-EDTA, an iron chelator that allows Tf recycling to take place in cell culture. Importantly, cRBCs produced by iPSC differentiation using the long PSC-RED protocol enucleate at much higher rates than with previous protocols, eliminating one of the impediments to the use of these cells to produce clinically useful cRBCs. The absence of albumin, the reduced amounts of Tf, the improved reproducibility associated with the elimination of all animal components, and the high yield on the RED protocols decrease the cost of production of cultured red blood cells. RED protocols should therefore help to make translational applications of cultured RBCs more economically realistic.


Assuntos
Diferenciação Celular , Eritrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Células Cultivadas , Eritrócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Leucócitos Mononucleares/citologia , Transferrina/farmacologia
6.
Curr Eye Res ; 25(3): 163-71, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12607186

RESUMO

PURPOSE: Although sickle (SS) red cell-mediated vaso-occlusion in retina and resultant retinopathy is well documented, the effects of SS red cells on choroidal vasculature are poorly understood. The intent of this study was to determine, using a rat model, the conditions under which retention of sickle erythrocytes in choroid occur and if that retention can be inhibited. METHODS: Sickle red cells were density separated into high density (SS4) or normal density, reticulocyte-enriched fractions (SS2). Red cells were labeled with FITC and administered IV to anesthetized Sprague Dawley rats. Rats were made either hypoxic or were given TNF-alpha intraperitoneally 5 hours before intravenous administration of red cells. Five minutes after administration of red cells, rats were exsanguinated, the retinas removed, and choroids prepared as flatmounts. The number of red cells retained in five high power fields of choroid was then determined. In other experiments, SS red cells were preincubated with the cyclic peptide TBC772 [inhibits binding of alpha4beta1 (VLA-4) and alpha4beta7 to their ligands], a control peptide TBC1194, or a VLA-4 neutralizing antibody before administration to the rat or antibodies against VLA-4 ligands were delivered IV before administration of SS red cells. RESULTS: Hypoxic conditions before administration of SS red cells significantly stimulated retention of SS4 cells (P = 0.0003), but did not significantly increase retention of SS2 cells. Administration of TNF-alpha significantly increased retention of all types of SS red cells (P < 0.001). Preincubation of cells with anti-VLA-4 or TBC 772 inhibited retention of SS red cells in choriocapillaris of TNF-alpha-treated rats (P < 0.0001). Complete inhibition of cytokine-stimulated retention was also accomplished by IV administration of monoclonal antibodies against fibronectin or its CS-1 domain, a ligand for VLA-4. CONCLUSIONS: The mechanisms for retention of SS red cells in retina and choroid appear identical: hypoxia-mediated retention of dense red cells and adherence of red cells in reticulocyte-rich fractions after cytokine stimulation. TNF-alpha-stimulated retention of SS red cells in choroid appears to be mediated by VLA-4, presumably on the surface of some reticulocytes. This increased retention was inhibited by a VLA-4 antagonist (TBC772), a VLA-4 neutralizing antibody or by blocking one of VLA-4's ligands, the CS-1 portion of fibronectin.


Assuntos
Anemia Falciforme/sangue , Corioide/irrigação sanguínea , Eritrócitos Anormais/metabolismo , Animais , Anticorpos Monoclonais , Adesão Celular , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Eritrócitos Anormais/efeitos dos fármacos , Fluoresceína-5-Isotiocianato , Humanos , Hipóxia/metabolismo , Integrina alfa4beta1/antagonistas & inibidores , Masculino , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley , Vasos Retinianos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
7.
Nat Med ; 16(2): 177-82, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20098432

RESUMO

Individuals with beta-thalassemia develop progressive systemic iron overload, resulting in high morbidity and mortality. These complications are caused by labile plasma iron, which is taken up by parenchymal cells in a dysregulated manner; in contrast, erythropoiesis depends on transferrin-bound iron uptake via the transferrin receptor. We hypothesized that the ineffective erythropoiesis and anemia observed in beta-thalassemia might be ameliorated by increasing the amount of circulating transferrin. We tested the ability of transferrin injections to modulate iron metabolism and erythropoiesis in Hbb(th1/th1) mice, an experimental model of beta-thalassemia. Injected transferrin reversed or markedly improved the thalassemia phenotype in these mice. Specifically, transferrin injections normalized labile plasma iron concentrations, increased hepcidin expression, normalized red blood cell survival and increased hemoglobin production; this treatment concomitantly decreased reticulocytosis, erythropoietin abundance and splenomegaly. These results indicate that transferrin is a limiting factor contributing to anemia in these mice and suggest that transferrin therapy might be beneficial in human beta-thalassemia.


Assuntos
Transferrina/uso terapêutico , Talassemia beta/tratamento farmacológico , Animais , Apoptose , Sobrevivência Celular , Modelos Animais de Doenças , Eritropoetina/sangue , Hematopoese Extramedular , Camundongos , Esplenomegalia/tratamento farmacológico , Transferrina/administração & dosagem , Talassemia beta/sangue
10.
Exp Hematol ; 37(2): 172-83, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19059700

RESUMO

OBJECTIVE: Beta-thalassemia results from beta-globin gene mutations that lead to ineffective erythropoiesis, shortened red cell survival, and anemia. Patients with beta-thalassemia develop iron overload, despite which, hepcidin levels are low. This suggests that hepcidin regulation in beta-thalassemia is more sensitive to factors unrelated to iron state. Our preliminary data demonstrates that Hbb(th1/th1) mice, a model of beta-thalassemia intermedia, have lower bone marrow iron levels while levels in the liver and spleen are increased; the later account for the increased systemic iron burden in beta-thalassemia intermedia. We hypothesized that exogenous iron would improve anemia in beta-thalassemia intermedia despite systemic iron overload and further suppress hepcidin secondary to progressive expansion of erythroid precursors. MATERIALS AND METHODS: We investigate parameters involved in red cell production, precursor apoptosis, parenchymal iron distribution, and hepcidin expression in iron treated Hbb(th1/th1) mice. RESULTS: Exogenous iron results in an expansion of erythroid precursors in the liver and spleen, leading to an increase in the number of red cells, reticulocytes, and hemoglobin production. A decrease in hepcidin expression is also observed. CONCLUSIONS: These findings demonstrate for the first time that iron results in expansion of extramedullary erythropoiesis, which improves anemia and suggests that expansion of extramedullary erythropoiesis itself results in hepcidin suppression in beta-thalassemia intermedia.


Assuntos
Eritropoese/efeitos dos fármacos , Hematínicos/farmacologia , Hematopoese Extramedular/efeitos dos fármacos , Hemoglobinas/biossíntese , Complexo Ferro-Dextran/farmacologia , Talassemia beta/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Medula Óssea/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Células Precursoras Eritroides/metabolismo , Eritropoese/genética , Hematopoese Extramedular/genética , Hemoglobinas/genética , Hepcidinas , Ferro/metabolismo , Camundongos , Camundongos Knockout , Mutação , Talassemia beta/tratamento farmacológico , Talassemia beta/genética
11.
Magn Reson Med ; 51(3): 525-32, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15004794

RESUMO

Neurological complications such as stroke are known consequences of sickle cell disease (SCD). In order to improve methods for the evaluation of stroke risk in SCD, MRI was used to evaluate cerebrovascular function in transgenic mouse models of human SCD. It is hypothesized that oxygen-sensitive imaging in the brain will reveal areas of excess deoxygenation that are either at risk of or the result of vaso-occlusion. Arterial spin labeling (ASL) perfusion was performed in order to correlate BOLD results with microvascular cerebral blood flow. Upon comparison with control animals, there was a relative increase in BOLD hyperoxia response of 42-67% (P < 0.001) in the transgenic mice while cerebral blood flow during normoxia was reduced by 30-40% (P < 0.02). Hyperoxia caused cerebral blood flow to decrease in control mice, whereas blood flow increased in the sickle transgenic mice. These results indicate impairment in brain autoregulation in the sickle cell transgenic mice leading to increased cerebral deoxyhemoglobin. Increased deoxyhemoglobin coupled with reduced perfusion may further increase the risk of vaso-occlusion and stroke. This may reflect polymer reduction or reduced cell adhesion during hyperoxia. The MRI protocol is noninvasive and thus directly applicable to a clinical population.


Assuntos
Anemia Falciforme/fisiopatologia , Circulação Cerebrovascular/fisiologia , Hiperóxia/fisiopatologia , Imageamento por Ressonância Magnética , Animais , Encéfalo/metabolismo , Meios de Contraste , Modelos Animais de Doenças , Hemoglobinas/análise , Homeostase/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Microcirculação/fisiologia , Oxigênio/sangue , Fatores de Risco , Marcadores de Spin , Acidente Vascular Cerebral/fisiopatologia
12.
Blood ; 99(4): 1103-8, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11830454

RESUMO

Nitric oxide (NO), essential for maintaining vascular tone, is produced from arginine by nitric oxide synthase. Plasma arginine levels are low in sickle cell anemia, and it is reported here that low plasma arginine is also found in our sickle transgenic mouse model that expresses human alpha, human beta(S), and human beta(S-Antilles) and is homozygous for the mouse beta(major) deletion (S+S-Antilles). S+S-Antilles mice were supplemented with a 4-fold increase in arginine that was maintained for several months. Mean corpuscular hemoglobin concentration (MCHC) decreased and the percent high-density red cells was reduced. Deoxy K(+) efflux is characteristic of red cells in sickle cell disease and contributes to the disease process by increasing the MCHC and rendering the cells more susceptible to polymer formation. This flux versus the room air flux was reduced in S+S-Antilles red cells from an average value of 1.6 +/- 0.3 mmol per liter of red cells x minute (FU) in nonsupplemented mice to 0.9 +/- 0.3 FU (n = 4, P < .02, paired t test) in supplemented mice. In room air, V(max) of the Ca(++)-activated K(+) channel (Gardos) was reduced from 4.1 +/- 0.6 FU (off diet) to 2.6 +/- 0.4 FU (n = 7 and 8, P < .04, t test) in arginine-supplemented mice versus clotrimazole. In conclusion, the major mechanism by which arginine supplementation reduces red cell density (MCHC) in S+S-Antilles mice is by inhibiting the Ca(++)-activated K(+) channel.


Assuntos
Anemia Falciforme/tratamento farmacológico , Arginina/farmacologia , Eritrócitos/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Animais , Arginina/administração & dosagem , Arginina/sangue , Citrulina/sangue , Modelos Animais de Doenças , Contagem de Eritrócitos , Eritrócitos/citologia , Hemoglobinas/efeitos dos fármacos , Hemoglobinas/metabolismo , Humanos , Cinética , Camundongos , Camundongos Transgênicos , Oxigênio/farmacologia , Potássio/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo
13.
Blood ; 103(6): 2384-90, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14615383

RESUMO

Elevation of K-Cl cotransport in patients with homozygous hemoglobin (Hb) S or HbC increases red cell mean corpuscular hemoglobin concentration (MCHC) and contributes significantly to pathology. Elucidation of the origin of elevated K-Cl cotransport in red cells with mutant hemoglobins has been confounded by the concomitant presence of reticulocytes with high K-Cl cotransport. In red cells of control mice (C57BL), transgenic mice that express only human HbA, and transgenic mice that express both mouse globins and human HbS, volume stimulation is weak and insensitive to NO3- and dihydroindenyl-oxy-alkanoic acid (DIOA). DIOA and NO3- are inhibitors in all other mammalian red cells. In contrast, in knock-out mice expressing exclusively human hemoglobin HbC or HbS+ gamma, replacement of isotonic Cl- media by hypotonic Cl- resulted in strong volume stimulation and sensitivity to DIOA, okadaic acid, and NO3-. In summary, we find that HbC, under all conditions, and HbS+ gamma, in the absence of mouse globins, have significant quantitative and qualitative effects on K-Cl cotransport in mouse red cells and activate mouse K-Cl. We conclude that human globins are able to stimulate the activity and/or regulation of K-Cl cotransport in mouse red cells. These observations support the contention that HbS and HbC stimulate K-Cl cotransport in human red cells.


Assuntos
Cloretos/metabolismo , Eritrócitos/metabolismo , Hemoglobina C/genética , Hemoglobina Falciforme/genética , Potássio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Ácidos Carboxílicos/farmacologia , Inibidores Enzimáticos/farmacologia , Hemoglobina A/genética , Humanos , Soluções Hipotônicas/farmacologia , Indenos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Nitratos/farmacologia , Ácido Okadáico/farmacologia
14.
Blood Cells Mol Dis ; 33(3): 303-7, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15528149

RESUMO

Hemoglobin E (HbE, beta26 Glu-->Lys) is the most common abnormal Hb variant in the world, and found in greatest frequency in Southeast (SE) Asia. In the United States, HbE is the third most prevalent variant (after HbS and HbC); and its now increasing frequency is due to immigration from SE Asia. HbE homozygotes present a benign clinical picture, but when HbE is coupled with beta0-thalassemia or HbS, variably severe hemoglobinopathies arise. To date, there are no transgenic animal models of HbE-related diseases. We report here the creation of transgenic mice expressing human HbE as a step toward creating animal models for HbE-related diseases. The betaE mice exhibit red blood cell hypochromia and target cells consistent with those observed in human patients exhibiting HbE trait. Furthermore, the transgenic HbE hemolysates contain increased amounts of Hb oxidation products.


Assuntos
Regulação da Expressão Gênica/genética , Hemoglobina E/genética , Homozigoto , Talassemia beta/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Talassemia beta/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA