Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Ther ; 29(6): 2019-2029, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-33609733

RESUMO

Gene disruption via programmable, sequence-specific nucleases represents a promising gene therapy strategy in which the reduction of specific protein levels provides a therapeutic benefit. Proprotein convertase subtilisin/kexin type 9 (PCSK9), an antagonist of the low-density lipoprotein (LDL) receptor, is a suitable target for nuclease-mediated gene disruption as an approach to treat hypercholesterolemia. We sought to determine the long-term durability and safety of PCSK9 knockdown in non-human primate (NHP) liver by adeno-associated virus (AAV)-delivered meganuclease following our initial report on the feasibility of this strategy. Six previously treated NHPs and additional NHPs administered AAV-meganuclease in combination with corticosteroid treatment or an alternative AAV serotype were monitored for a period of up to 3 years. The treated NHPs exhibited a sustained reduction in circulating PCSK9 and LDL cholesterol (LDL-c) through the course of the study concomitant with stable gene editing of the PCSK9 locus. Low-frequency off-target editing remained stable, and no obvious adverse changes in histopathology of the liver were detected. We demonstrate similar on-target nuclease activity in primary human hepatocytes using a chimeric liver-humanized mouse model. These studies demonstrate that targeted in vivo gene disruption exerts a lasting therapeutic effect and provide pivotal data for safety considerations, which support clinical translation.


Assuntos
Edição de Genes , Lipoproteínas LDL/metabolismo , Pró-Proteína Convertase 9/genética , Animais , Sistemas CRISPR-Cas , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Lipoproteínas LDL/genética , Fígado/metabolismo , Camundongos , Camundongos Knockout , Primatas , Pró-Proteína Convertase 9/metabolismo
2.
Blood ; 133(26): 2745-2752, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-30975639

RESUMO

Many genetic diseases, including hemophilia, require long-term therapeutic effects. Despite the initial success of liver-directed adeno-associated virus (AAV) gene therapy for hemophilia in clinical trials, long-term sustained therapeutic effects have yet to be seen. One explanation for the gradual decline of efficacy over time is that the nonintegrating AAV vector genome could be lost during cell division during hepatocyte turnover, albeit at a slow pace in adults. Readministering the same vector is challenging as a result of the AAV-neutralizing antibodies elicited by the initial treatment. Here, we investigated the use of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated homology-directed gene targeting for sustained treatment of hemophilia B. We developed a donor vector containing a promoterless partial human factor IX (FIX) complementary DNA carrying the hyperactive FIX Padua mutation. A single injection of dual AAV vectors in newborn and adult FIX-knockout (FIX-KO) mice led to stable expression of FIX at or above the normal levels for 8 months. Eight weeks after the vector treatment, we subjected a subgroup of newborn and adult treated FIX-KO mice to a two-thirds partial hepatectomy; all of these animals survived the procedure without any complications or interventions. FIX levels persisted at similar levels for 24 weeks after partial hepatectomy, indicating stable genomic targeting. Our results lend support for the use of a CRISPR/Cas9 approach to achieve lifelong expression of therapeutic proteins.


Assuntos
Sistemas CRISPR-Cas , Fator IX/genética , Marcação de Genes/métodos , Hemofilia B/genética , Hemostasia/genética , Animais , Animais Recém-Nascidos , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Humanos , Camundongos , Camundongos Knockout
3.
Mol Genet Metab ; 120(4): 299-305, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28283349

RESUMO

Ornithine transcarbamylase (OTC) deficiency is an X-linked disorder of the urea cycle. Hemizygous males and heterozygous females may experience life-threatening elevations of ammonia in blood and brain, leading to irreversible cognitive impairment, coma, and death. Recent evidence of acute liver failure and fibrosis/cirrhosis is also emerging in OTC-deficient patients. Here, we investigated the long-term consequences of abnormal ureagenesis in female mice heterozygous (Het) for a null mutation in the OTC gene. Two-month-old Het OTC knockout (KO) mice received a single dose of self-complementary adeno-associated virus (AAV) encoding a codon-optimized human OTC gene at 1×1010, 3×1010, or 1×1011 vector genome copies per mouse. We compared liver pathology from 18-month-old treated Het OTC-KO mice, age-matched untreated Het OTC-KO mice, and WT littermates, and assessed urinary orotic acid levels and vector genome copies in liver at 4, 10, and 16months following vector administration. Het OTC-KO female mice showed evidence of liver inflammation and the eventual development of significant fibrosis. Treatment with AAV gene therapy not only corrected the underlying metabolic abnormalities, but also prevented the development of liver fibrosis. Our study demonstrates that early treatment of OTC deficiency with gene therapy may prevent clinically relevant consequences of chronic liver damage from developing.


Assuntos
Envelhecimento/genética , Vetores Genéticos/administração & dosagem , Cirrose Hepática/prevenção & controle , Doença da Deficiência de Ornitina Carbomoiltransferase/terapia , Ornitina Carbamoiltransferase/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Terapia Genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Doença da Deficiência de Ornitina Carbomoiltransferase/complicações , Doença da Deficiência de Ornitina Carbomoiltransferase/genética , Resultado do Tratamento
4.
Mol Ther ; 23(12): 1877-87, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26412589

RESUMO

Vectors based on the clade E family member adeno-associated virus (AAV) serotype 8 have shown promise in patients with hemophilia B and have emerged as best in class for human liver gene therapies. We conducted a thorough evaluation of liver-directed gene therapy using vectors based on several natural and engineered capsids including the clade E AAVrh10 and the largely uncharacterized and phylogenically distinct AAV3B. Included in this study was a putatively superior hepatotropic capsid, AAVLK03, which is very similar to AAV3B. Vectors based on these capsids were benchmarked against AAV8 and AAV2 in a number of in vitro and in vivo model systems including C57BL/6 mice, immune-deficient mice that are partially repopulated with human hepatocytes, and nonhuman primates. Our studies in nonhuman primates and human hepatocytes demonstrated high level transduction of the clade E-derived vectors and equally high transduction with vectors based on AAV3B. In contrast to previous reports, AAVLK03 vectors are not superior to either AAV3B or AAV8. Vectors based on AAV3B should be considered for liver-directed gene therapy when administered following, or before, treatment with the serologically distinct clade E vectors.


Assuntos
Proteínas do Capsídeo/genética , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Fígado/citologia , Animais , Proteínas do Capsídeo/metabolismo , Engenharia Genética , Terapia Genética , Hepatócitos/metabolismo , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução Genética
5.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 10): 2061-71, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24100324

RESUMO

A variant Hb ζ2ß2(s) that is formed from sickle hemoglobin (Hb S; α2ß2(s)) by exchanging adult α-globin with embryonic ζ-globin subunits shows promise as a therapeutic agent for sickle-cell disease (SCD). Hb ζ2ß2(s) inhibits the polymerization of deoxygenated Hb S in vitro and reverses characteristic features of SCD in vivo in mouse models of the disorder. When compared with either Hb S or with normal human adult Hb A (α2ß2), Hb ζ2ß2(s) exhibits atypical properties that include a high oxygen affinity, reduced cooperativity, a weak Bohr effect and blunted 2,3-diphosphoglycerate allostery. Here, the 1.95 Šresolution crystal structure of human Hb ζ2ß2(s) that was expressed in complex transgenic knockout mice and purified from their erythrocytes is presented. When fully liganded with carbon monoxide, Hb ζ2ß2(s) displays a central water cavity, a ζ1-ß(s)2 (or ζ2-ß(s)1) interface, intersubunit salt-bridge/hydrogen-bond interactions, C-terminal ßHis146 salt-bridge interactions, and a ß-cleft, that are highly unusual for a relaxed hemoglobin structure and are more typical of a tense conformation. These quaternary tense-like features contrast with the tertiary relaxed-like conformations of the ζ1ß(s)1 dimer and the CD and FG corners, as well as the overall structures of the heme cavities. This crystallographic study provides insights into the altered oxygen-transport properties of Hb ζ2ß2(s) and, moreover, decouples tertiary- and quaternary-structural events that are critical to Hb ligand binding and allosteric function.


Assuntos
Anemia Falciforme/sangue , Anemia Falciforme/genética , Hemoglobina Falciforme/genética , Globinas zeta/química , Adulto , Regulação Alostérica/genética , Animais , Cristalografia por Raios X , Variação Genética , Hemoglobina Falciforme/metabolismo , Humanos , Ligantes , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Oxigênio/metabolismo , Ligação Proteica/genética , Conformação Proteica , Multimerização Proteica/genética , alfa-Globinas/genética , alfa-Globinas/metabolismo , Globinas zeta/metabolismo
6.
Nat Biotechnol ; 2023 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-37932420

RESUMO

The development of liver-based adeno-associated virus (AAV) gene therapies is facing concerns about limited efficiency and durability of transgene expression. We evaluated nonhuman primates following intravenous dosing of AAV8 and AAVrh10 vectors for over 2 years to better define the mechanism(s) of transduction that affect performance. High transduction of non-immunogenic transgenes was achieved, although expression declined over the first 90 days to reach a lower but stable steady state. More than 10% of hepatocytes contained single nuclear domains of vector DNA that persisted despite the loss of transgene expression. Greater reductions in vector DNA and RNA were observed with immunogenic transgenes. Genomic integration of vector sequences, including complex concatemeric structures, were detected in 1 out of 100 cells at broadly distributed loci that were not in proximity to genes associated with hepatocellular carcinoma. Our studies suggest that AAV-mediated transgene expression in primate hepatocytes occurs in two phases: high but short-lived expression from episomal genomes, followed by much lower but stable expression, likely from integrated vectors.

7.
Mol Ther Methods Clin Dev ; 24: 292-305, 2022 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-35211641

RESUMO

Ornithine transcarbamylase deficiency is a rare X-linked genetic urea cycle disorder leading to episodes of acute hyperammonemia, adverse cognitive and neurological effects, hospitalizations, and in some cases death. DTX301, a non-replicating, recombinant self-complimentary adeno-associated virus vector serotype 8 (scAAV8)-encoding human ornithine transcarbamylase, is a promising gene therapy for ornithine transcarbamylase deficiency; however, the impact of sex and prophylactic immunosuppression on ornithine transcarbamylase gene therapy outcomes is not well characterized. This study sought to describe the impact of sex and immunosuppression in adult, sexually mature female and male cynomolgus macaques through day 140 after DTX301 administration. Four study groups (n = 3/group) were included: male non-immunosuppressed; male immunosuppressed; female non-immunosuppressed; and female immunosuppressed. DTX301 was well tolerated with and without immunosuppression; no notable differences were observed between female and male groups across outcome measures. Prednisolone-treated animals exhibited a trend toward greater vector genome and transgene expression, although the differences were not statistically significant. The hepatic interferon gene signature was significantly decreased in prednisolone-treated animals, and a significant inverse relationship was observed between interferon gene signature levels and hepatic vector DNA and transgene RNA. These observations were not sustained upon immunosuppression withdrawal. Further studies may determine whether the observed effect can be prolonged.

8.
Nat Med ; 10(4): 365-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15034572

RESUMO

Gene replacement or gene reactivation therapies for sickle-cell disease (SCD) typically target the mutant beta(S)-globin subunits of hemoglobin-S (alpha(2)beta(S)(2)) for substitution by nonpathological beta-like globins. Here we show, in vitro and in vivo in a transgenic mouse model of SCD, that the adverse properties of hemoglobin-S can be reversed by exchanging its normal alpha-globin subunits for zeta-globin, an endogenous, developmentally silenced, non-beta-like globin.


Assuntos
Antidrepanocíticos/uso terapêutico , Globinas/uso terapêutico , Animais , Cromatografia Líquida de Alta Pressão , Globinas/fisiologia , Humanos , Camundongos , Camundongos Transgênicos
9.
Mol Ther Methods Clin Dev ; 22: 1-10, 2021 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-34258325

RESUMO

Gene therapy for hypercholesterolemia offers the potential to sustainably ameliorate disease for life with a single dose. In this study, we demonstrate the combinatorial effects of codon and vector optimization, which significantly improve the efficacy of an adeno-associated virus (AAV) vector in the low-density lipoprotein receptor (LDLR)-deficient mouse model (Ldlr -/-, Apobec1 -/- double knockout [DKO]). This study investigated vector efficacy following the combination of intervening sequence 2 (IVS2) of the human beta-globin gene and codon optimization with the previously developed gain-of-function, human LDLR triple-mutant variant (hLDLR-L318D/K809R/C818A) in the treatment of homozygous familial hypercholesterolemia (HoFH). Vector doses as low as 3 × 1011 genome copies (GC)/kg achieved a robust reduction of serum low-density lipoprotein cholesterol (LDL-C) by 98% in male LDLR-deficient mice. Less efficient LDL-C reduction was observed in female mice, which was attributable to lower gene transfer efficiency in liver. We also observed persistent and stable transgene expression for 120 days, with LDL-C levels being undetectable in male DKO mice treated with the second-generation vector. In conclusion, codon and vector optimization enhanced transgene expression and reduced serum LDL-C levels effectively at a lower dose in LDLR-deficient mice. The second-generation clinical candidate vector we have developed has the potential to achieve therapeutic effects in HoFH patients.

10.
Sci Adv ; 6(7): eaax5701, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32095520

RESUMO

Ornithine transcarbamylase (OTC) deficiency is an X-linked urea cycle disorder associated with high mortality. Although a promising treatment for late-onset OTC deficiency, adeno-associated virus (AAV) neonatal gene therapy would only provide short-term therapeutic effects as the non-integrated genome gets lost during hepatocyte proliferation. CRISPR-Cas9-mediated homology-directed repair can correct a G-to-A mutation in 10% of OTC alleles in the livers of newborn OTC spfash mice. However, an editing vector able to correct one mutation would not be applicable for patients carrying different OTC mutations, plus expression would not be fast enough to treat a hyperammonemia crisis. Here, we describe a dual-AAV vector system that accomplishes rapid short-term expression from a non-integrated minigene and long-term expression from the site-specific integration of this minigene without any selective growth advantage for OTC-positive cells in newborns. This CRISPR-Cas9 gene-targeting approach may be applicable to all patients with OTC deficiency, irrespective of mutation and/or clinical state.


Assuntos
Sistemas CRISPR-Cas/genética , Marcação de Genes , Terapia Genética , Mutação/genética , Doença da Deficiência de Ornitina Carbomoiltransferase/genética , Doença da Deficiência de Ornitina Carbomoiltransferase/terapia , Animais , Reparo do DNA/genética , Dependovirus/genética , Proteínas Alimentares , Modelos Animais de Doenças , Loci Gênicos , Vetores Genéticos/metabolismo , Mutação INDEL/genética , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Ornitina Carbamoiltransferase/genética , Ornitina Carbamoiltransferase/metabolismo , Fatores de Tempo
11.
Nat Biotechnol ; 36(8): 717-725, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985478

RESUMO

Clinical translation of in vivo genome editing to treat human genetic diseases requires thorough preclinical studies in relevant animal models to assess safety and efficacy. A promising approach to treat hypercholesterolemia is inactivating the secreted protein PCSK9, an antagonist of the LDL receptor. Here we show that single infusions in six non-human primates of adeno-associated virus vector expressing an engineered meganuclease targeting PCSK9 results in dose-dependent disruption of PCSK9 in liver, as well as a stable reduction in circulating PCSK9 and serum cholesterol. Animals experienced transient, asymptomatic elevations of serum transaminases owing to the formation of T cells against the transgene product. Vector DNA and meganuclease expression declined rapidly, leaving stable populations of genome-edited hepatocytes. A second-generation PCSK9-specific meganuclease showed reduced off-target cleavage. These studies demonstrate efficient, physiologically relevant in vivo editing in non-human primates, and highlight safety considerations for clinical translation.


Assuntos
Colesterol/sangue , Desoxirribonucleases/metabolismo , Fígado/enzimologia , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Animais , Dependovirus/genética , Edição de Genes , Vetores Genéticos , Células HEK293 , Hepatócitos/metabolismo , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/terapia , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Knockout , Receptores de LDL/antagonistas & inibidores
12.
Nat Biotechnol ; 34(3): 334-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26829317

RESUMO

Many genetic liver diseases in newborns cause repeated, often lethal, metabolic crises. Gene therapy using nonintegrating viruses such as adeno-associated virus (AAV) is not optimal in this setting because the nonintegrating genome is lost as developing hepatocytes proliferate. We reasoned that newborn liver may be an ideal setting for AAV-mediated gene correction using CRISPR-Cas9. Here we intravenously infuse two AAVs, one expressing Cas9 and the other expressing a guide RNA and the donor DNA, into newborn mice with a partial deficiency in the urea cycle disorder enzyme, ornithine transcarbamylase (OTC). This resulted in reversion of the mutation in 10% (6.7-20.1%) of hepatocytes and increased survival in mice challenged with a high-protein diet, which exacerbates disease. Gene correction in adult OTC-deficient mice was lower and accompanied by larger deletions that ablated residual expression from the endogenous OTC gene, leading to diminished protein tolerance and lethal hyperammonemia on a chow diet.


Assuntos
Sistemas CRISPR-Cas/genética , Doença da Deficiência de Ornitina Carbomoiltransferase/terapia , Ornitina Carbamoiltransferase/genética , Edição de RNA , Animais , Dependovirus/genética , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Ornitina Carbamoiltransferase/uso terapêutico , Doença da Deficiência de Ornitina Carbomoiltransferase/genética , Vírus/genética
13.
Hum Gene Ther Methods ; 27(6): 228-237, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27903094

RESUMO

Numerous methods of vector design and delivery have been employed in an attempt to increase transgene expression following AAV-based gene therapy. Here, a gene transfer study was conducted in mice to compare the effects of vector self-complementarity (double- or single-stranded DNA), codon optimization of the transgene, and vector dose on transgene expression levels in the liver. Two different reporter genes were used: human ornithine transcarbamylase (hOTC) detected by immunofluorescence, and enhanced green fluorescent protein (EGFP) detected by direct fluorescence. The AAV8 capsid was chosen for all experiments due to its strong liver tropism. While EGFP is already a codon-optimized version of the original gene, both wild-type (WT) and codon-optimized (co) versions of the hOTC transgene were compared in this study. In addition, the study evaluated which of the two hOTC modifications-codon optimization or self-complementarity-would confer the highest increase in expression levels at a given dose. Interestingly, based on morphometric image analysis, it was observed that the difference in detectable expression levels between self-complementary (sc) and single-stranded (ss) hOTCco vectors was dose dependent, with a sevenfold increase in OTC-positive area using sc vectors at a dose of 3 × 109 genome copies (GC) per mouse, but no significant difference at a dose of 1 × 1010 GC/mouse. In contrast, with EGFP as a transgene, the increases in expression levels when using the sc vector were observed at both the 3 × 109 GC/mouse and 1 × 1010 GC/mouse doses. Furthermore, codon optimization of the hOTC transgene generated a more significant improvement in expression than the use of self-complementarity did. Overall, the results demonstrate that increases in expression levels gained by using sc vectors instead of ss vectors can vary between different transgenes, and that codon optimization of the transgene can have an even more powerful effect on the resulting expression levels.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Transdução Genética , Animais , Códon , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/uso terapêutico , Humanos , Fígado/metabolismo , Camundongos , Ornitina Carbamoiltransferase/genética , Ornitina Carbamoiltransferase/uso terapêutico
14.
J Hematol Oncol ; 7: 35, 2014 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-24751163

RESUMO

BACKGROUND: The normal accumulation of adult α and ß globins in definitive erythrocytes is critically dependent upon processes that ensure that the cognate mRNAs are maintained at high levels in transcriptionally silent, but translationally active progenitor cells. The impact of these post-transcriptional regulatory events on the expression of embryonic ζ globin is not known, as its encoding mRNA is not normally transcribed during adult erythropoiesis. Recently, though, ζ globin has been recognized as a potential therapeutic for α thalassemia and sickle-cell disease, raising practical questions about constitutive post-transcriptional processes that may enhance, or possibly prohibit, the expression of exogenous or derepresssed endogenous ζ-globin genes in definitive erythroid progenitors. METHODS: The present study assesses mRNA half-life in intact cells using a pulse-chase approach; identifies cis-acting determinants of ζ-globin mRNA stability using a saturation mutagenesis strategy; establishes critical 3'UTR secondary structures using an in vitro enzymatic mapping method; and identifies trans-acting effector factors using an affinity chromatographical procedure. RESULTS: We specify a tetranucleotide 3'UTR motif that is required for the high-level accumulation of ζ-globin transcripts in cultured cells, and formally demonstrate that it prolongs their cytoplasmic half-lives. Surprisingly, the ζ-globin mRNA stability motif does not function autonomously, predicting an activity that is subject to structural constraints that we subsequently specify. Additional studies demonstrate that the ζ-globin mRNA stability motif is targeted by AUF1, a ubiquitous RNA-binding protein that enhances the half-life of adult ß-globin mRNA, suggesting commonalities in post-transcriptional processes that regulate globin transcripts at all stages of mammalian development. CONCLUSIONS: These data demonstrate a mechanism for ζ-globin mRNA stability that exists in definitive erythropoiesis and is available for therapeutic manipulation in α thalassemia and sickle-cell disease.


Assuntos
Regiões 3' não Traduzidas/genética , Estabilidade de RNA , RNA Mensageiro/genética , Globinas zeta/genética , Adulto , Sequência de Bases , Citoplasma/genética , Citoplasma/metabolismo , Expressão Gênica , Meia-Vida , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Humanos , Immunoblotting , Dados de Sequência Molecular , Mutação , Conformação de Ácido Nucleico , Motivos de Nucleotídeos/genética , Ligação Proteica , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico
15.
Blood ; 109(2): 795-801, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17003365

RESUMO

Functional studies of embryonic epsilon-globin indicate that individuals with beta thalassemia or sickle cell disease are likely to benefit from therapeutic, transcriptional derepression of its encoding gene. The success of epsilon-globin gene-reactivation strategies, however, will be tempered by the stability that epsilon-globin mRNA exhibits in developmental stage-discordant definitive erythroid progenitors. Using cell culture and transgenic mouse model systems, we demonstrate that epsilon-globin mRNA is modestly unstable in immature, transcriptionally active erythroid cells, but that this characteristic has relatively little impact on the accumulation of epsilon-globin mRNA at subsequent stages of terminal differentiation. Importantly, the constitutive stability of epsilon-globin mRNA increases in transgenic mouse models of beta thalassemia, suggesting that epsilon- and beta-globin mRNAs are coregulated through a shared posttranscriptional mechanism. As anticipated, relevant cis-acting determinants of epsilon-globin mRNA stability map to its 3' UTR, consistent with the positioning of functionally related elements in other globin mRNAs. These studies demonstrate that posttranscriptional processes do not pose a significant practical barrier to epsilon-globin gene reactivation and, moreover, indicate that related therapeutic strategies may be particularly effective in individuals carrying beta-thalassemic gene defects.


Assuntos
Regulação da Expressão Gênica , Globinas/genética , Interferência de RNA , Transcrição Gênica , Talassemia beta/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Globinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , RNA Mensageiro/genética
16.
Biochem Biophys Res Commun ; 325(4): 1376-82, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15555579

RESUMO

Hemoglobin zeta(2)beta(2)(S) is generated by substituting embryonic zeta-globin subunits for the normal alpha-globin components of Hb S (alpha(2)beta(2)(S)). This novel hemoglobin has recently been shown to inhibit polymerization of Hb S in vitro and to normalize the pathological phenotype of mouse models of sickle cell disease in vivo. Despite its promise as a therapeutic tool in human disease, however, the basic O(2)-transport properties of Hb zeta(2)beta(2)(S) have not yet been described. Using human hemoglobins purified from complex transgenic-knockout mice, we show that Hb zeta(2)beta(2)(S) exhibits an O(2) affinity as well as a Hill coefficient, Bohr response, and allosteric properties in vitro that are suboptimally suited for physiological O(2) transport in vivo. These data are substantiated by in situ analyses demonstrating an increase in the O(2) affinity of intact erythrocytes from mice that express Hb zeta(2)beta(2)(S). Surprisingly, though, co-expression of Hb zeta(2)beta(2)(S) leads to a substantial improvement in the tissue oxygenation of mice that model sickle cell disease. These analyses suggest that, in the context of sickle cell disease, the beneficial antisickling effects of Hb zeta(2)beta(2)(S) outweigh its O(2)-transport liabilities. The potential structural bases for the antisickling properties of Hb zeta(2)beta(2)(S) are discussed in the context of these new observations.


Assuntos
Anemia Falciforme/metabolismo , Globinas/química , Globinas/metabolismo , Hemoglobina Falciforme/química , Hemoglobina Falciforme/metabolismo , Oxigênio/química , Oxigênio/metabolismo , Animais , Sítios de Ligação , Transporte Biológico Ativo/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Cinética , Camundongos , Camundongos Transgênicos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
17.
Proc Natl Acad Sci U S A ; 99(16): 10635-40, 2002 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-12124399

RESUMO

The principle that developmentally silenced globin genes can be reactivated in adults with defects in beta-globin gene expression has been well established both in vitro and in vivo. In practice, levels of developmental stage-discordant fetal gamma globin that can be achieved by using currently approved therapies are generally insufficient to fully resolve typical clincopathological features of sickle cell disease. The therapeutic potential of another developmentally silenced globin--embryonic epsilon globin--has been difficult to evaluate in the absence of a convenient expression system or an appropriate experimental model. The current work analyzes the antisickling properties of an epsilon -globin-containing heterotetramer (Hb Gower-2) both in vitro as well as in vivo in a well-established mouse model of sickle cell anemia. These animals, expressing 100% human Hb S, display a chronic hemolytic anemia with compensatory marrow and extramedullary erythropoiesis, abundant circulating sickled erythrocytes, and chronic tissue damage evidenced by parallel histopathological and functional deficits. By comparison, related mice that coexpress Hb S as well as Hb Gower-2 exhibit normal physiological, morphological, histological, and functional attributes. Subsequent in vitro analyses substantiate results from whole-animal studies, indicating that the polymerization of deoxygenated Hb S can be significantly slowed by relatively small quantities of Hb Gower-2. Together, the in vivo and in vitro analyses suggest that reactivation of epsilon-globin gene expression would be therapeutically beneficial to adults with sickle phenotypes, and provide a rationale for detailed investigations into the molecular basis for its developmental silencing.


Assuntos
Anemia Falciforme/patologia , Globinas/fisiologia , Hemoglobina Falciforme/fisiologia , Hemoglobinas Anormais/fisiologia , Anemia Falciforme/fisiopatologia , Animais , Modelos Animais de Doenças , Eritrócitos , Expressão Gênica , Globinas/genética , Hematopoese Extramedular , Doença da Hemoglobina SC/patologia , Doença da Hemoglobina SC/fisiopatologia , Hemoglobina Falciforme/genética , Hemoglobinas Anormais/genética , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Polímeros , Síndrome , Talassemia beta/patologia , Talassemia beta/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA