Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Elife ; 122024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38885133

RESUMO

Despite the central role of T cells in tumor immunity, attempts to harness their cytotoxic capacity as a therapy have met limited efficacy, partially as a result of the suppressive microenvironment which limits their migration and activation. In contrast, myeloid cells massively infiltrate tumors and are well adapted to survive these harsh conditions. While they are equipped with cell-killing abilities, they often adopt an immunosuppressive phenotype upon migration to tumors. Therefore, the questions of how to modify their activation programming against cancer, and what signaling cascades should be activated in myeloid cells to elicit their cytotoxicity have remained unclear. Here, we found that activation of IgM-induced signaling in murine myeloid cells results in secretion of lytic granules and massive tumor cell death. These findings open venues for designing novel immunotherapy by equipping monocytes with chimeric receptors that target tumor antigens and consequently, signal through IgM receptor. Nonetheless, we found that myeloid cells do not express the antibody-derived portion used to recognize the tumor antigen due to the induction of an ER stress response. To overcome this limitation, we designed chimeric receptors that are based on the high-affinity FcγRI for IgG. Incubation of macrophages expressing these receptors along with tumor-binding IgG induced massive tumor cell killing and secretion of reactive oxygen species and Granzyme B. Overall, this work highlights the challenges involved in genetically reprogramming the signaling in myeloid cells and provides a framework for endowing myeloid cells with antigen-specific cytotoxicity.


Assuntos
Células Mieloides , Receptores de IgG , Animais , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Camundongos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Imunoglobulina M/metabolismo , Imunoglobulina M/imunologia , Transdução de Sinais , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias/imunologia
3.
JCI Insight ; 7(17)2022 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-35980743

RESUMO

Development of resistance to chemo- and immunotherapies often occurs following treatment of melanoma brain metastasis (MBM). The brain microenvironment (BME), particularly astrocytes, cooperate toward MBM progression by upregulating secreted factors, among which we found that monocyte chemoattractant protein-1 (MCP-1) and its receptors, CCR2 and CCR4, were overexpressed in MBM compared with primary lesions. Among other sources of MCP-1 in the brain, we show that melanoma cells altered astrocyte secretome and evoked MCP-1 expression and secretion, which in turn induced CCR2 expression in melanoma cells, enhancing in vitro tumorigenic properties, such as proliferation, migration, and invasion of melanoma cells. In vivo pharmacological blockade of MCP-1 or molecular knockout of CCR2/CCR4 increased the infiltration of cytotoxic CD8+ T cells and attenuated the immunosuppressive phenotype of the BME as shown by decreased infiltration of Tregs and tumor-associated macrophages/microglia in several models of intracranially injected MBM. These in vivo strategies led to decreased MBM outgrowth and prolonged the overall survival of the mice. Our findings highlight the therapeutic potential of inhibiting interactions between BME and melanoma cells for the treatment of this disease.


Assuntos
Neoplasias Encefálicas , Melanoma , Animais , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Quimiocina CCL2/metabolismo , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Receptores CCR2/metabolismo , Microambiente Tumoral
4.
Nat Biotechnol ; 40(8): 1241-1249, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35681059

RESUMO

Transplantation of B cells engineered ex vivo to secrete broadly neutralizing antibodies (bNAbs) has shown efficacy in disease models. However, clinical translation of this approach would require specialized medical centers, technically demanding protocols and major histocompatibility complex compatibility of donor cells and recipients. Here we report in vivo B cell engineering using two adeno-associated viral vectors, with one coding for Staphylococcus aureus Cas9 (saCas9) and the other for 3BNC117, an anti-HIV bNAb. After intravenously injecting the vectors into mice, we observe successful editing of B cells leading to memory retention and bNAb secretion at neutralizing titers of up to 6.8 µg ml-1. We observed minimal clustered regularly interspaced palindromic repeats (CRISPR)-Cas9 off-target cleavage as detected by unbiased CHANGE-sequencing analysis, whereas on-target cleavage in undesired tissues is reduced by expressing saCas9 from a B cell-specific promoter. In vivo B cell engineering to express therapeutic antibodies is a safe, potent and scalable method, which may be applicable not only to infectious diseases but also in the treatment of noncommunicable conditions, such as cancer and autoimmune disease.


Assuntos
Infecções por HIV , HIV-1 , Animais , Anticorpos Neutralizantes/genética , Linfócitos B , Anticorpos Amplamente Neutralizantes , Anticorpos Anti-HIV/genética , Infecções por HIV/terapia , Camundongos , Staphylococcus aureus
5.
Nat Biotechnol ; 40(12): 1807-1813, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35773341

RESUMO

Multiple clinical trials of allogeneic T cell therapy use site-specific nucleases to disrupt T cell receptor (TCR) and other genes1-6. In this study, using single-cell RNA sequencing, we investigated genome editing outcomes in primary human T cells transfected with CRISPR-Cas9 and guide RNAs targeting genes for TCR chains and programmed cell death protein 1. Four days after transfection, we found a loss of chromosome 14, harboring the TCRα locus, in up to 9% of the cells and a chromosome 14 gain in up to 1.4% of the cells. Chromosome 7, harboring the TCRß locus, was truncated in 9.9% of the cells. Aberrations were validated using fluorescence in situ hybridization and digital droplet PCR. Aneuploidy was associated with reduced proliferation, induced p53 activation and cell death. However, at 11 days after transfection, 0.9% of T cells still had a chromosome 14 loss. Aneuploidy and chromosomal truncations are, thus, frequent outcomes of CRISPR-Cas9 cleavage that should be monitored and minimized in clinical protocols.


Assuntos
Sistemas CRISPR-Cas , Linfócitos T , Humanos , Sistemas CRISPR-Cas/genética , Hibridização in Situ Fluorescente , Edição de Genes/métodos , Receptores de Antígenos de Linfócitos T/genética , Aneuploidia
6.
Front Genome Ed ; 4: 785698, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35359664

RESUMO

Many inborn errors of metabolism require life-long treatments and, in severe conditions involving the liver, organ transplantation remains the only curative treatment. Non-integrative AAV-mediated gene therapy has shown efficacy in adult patients. However, treatment in pediatric or juvenile settings, or in conditions associated with hepatocyte proliferation, may result in rapid loss of episomal viral DNA and thus therapeutic efficacy. Re-administration of the therapeutic vector later in time may not be possible due to the presence of anti-AAV neutralizing antibodies. We have previously shown the permanent rescue of the neonatal lethality of a Crigler-Najjar mouse model by applying an integrative gene-therapy based approach. Here, we targeted the human coagulation factor IX (hFIX) cDNA into a hemophilia B mouse model. Two AAV8 vectors were used: a promoterless vector with two arms of homology for the albumin locus, and a vector carrying the CRISPR/SaCas9 and the sgRNA. Treatment of neonatal P2 wild-type mice resulted in supraphysiological levels of hFIX being stable 10 months after dosing. A single injection of the AAV vectors into neonatal FIX KO mice also resulted in the stable expression of above-normal levels of hFIX, reaching up to 150% of the human levels. Mice subjected to tail clip analysis showed a clotting capacity comparable to wild-type animals, thus demonstrating the rescue of the disease phenotype. Immunohistological analysis revealed clusters of hFIX-positive hepatocytes. When we tested the approach in adult FIX KO mice, we detected hFIX in plasma by ELISA and in the liver by western blot. However, the hFIX levels were not sufficient to significantly ameliorate the bleeding phenotype upon tail clip assay. Experiments conducted using a AAV donor vectors containing the eGFP or the hFIX cDNAs showed a higher recombination rate in P2 mice compared to adult animals. With this study, we demonstrate an alternative gene targeting strategy exploiting the use of the CRISPR/SaCas9 platform that can be potentially applied in the treatment of pediatric patients suffering from hemophilia, also supporting its application to other liver monogenic diseases. For the treatment of adult patients, further studies for the improvement of targeting efficiency are still required.

7.
Front Immunol ; 12: 705381, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34349765

RESUMO

The role of B cells in the tumor microenvironment (TME) has largely been under investigated, and data regarding the antibody repertoire encoded by B cells in the TME and the adjacent lymphoid organs are scarce. Here, we utilized B cell receptor high-throughput sequencing (BCR-Seq) to profile the antibody repertoire signature of tumor-infiltrating lymphocyte B cells (TIL-Bs) in comparison to B cells from three anatomic compartments in a mouse model of triple-negative breast cancer. We found that TIL-Bs exhibit distinct antibody repertoire measures, including high clonal polarization and elevated somatic hypermutation rates, suggesting a local antigen-driven B-cell response. Importantly, TIL-Bs were highly mutated but non-class switched, suggesting that class-switch recombination may be inhibited in the TME. Tracing the distribution of TIL-B clones across various compartments indicated that they migrate to and from the TME. The data thus suggests that antibody repertoire signatures can serve as indicators for identifying tumor-reactive B cells.


Assuntos
Diversidade de Anticorpos , Subpopulações de Linfócitos B/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Experimentais/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Neoplasias de Mama Triplo Negativas/imunologia , Microambiente Tumoral/imunologia , Animais , Células Sanguíneas/imunologia , Medula Óssea/patologia , Linhagem Celular Tumoral/transplante , Movimento Celular , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Região Variável de Imunoglobulina/genética , Linfonodos/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Receptores de Antígenos de Linfócitos B/genética , Hipermutação Somática de Imunoglobulina , Neoplasias de Mama Triplo Negativas/patologia
8.
Hepatology ; 73(6): 2223-2237, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32976669

RESUMO

BACKGROUND AND AIMS: Adeno-associated viral (AAV) gene therapy has shown great promise as an alternative treatment for metabolic disorders managed using liver transplantation, but remains limited by transgene loss and genotoxicity. Our study aims to test an AAV vector with a promoterless integrating cassette, designed to provide sustained hepatic transgene expression and reduced toxicity in comparison to canonical AAV therapy. APPROACH AND RESULTS: Our AAV vector was designed to insert a methylmalonyl-CoA mutase (MMUT) transgene into the 3' end of the albumin locus and tested in mouse models of methylmalonic acidemia (MMA). After neonatal delivery, we longitudinally evaluated hepatic transgene expression, plasma levels of methylmalonate, and the MMA biomarker, fibroblast growth factor 21 (Fgf21), as well as integration of MMUT in the albumin locus. At necropsy, we surveyed for AAV-related hepatocellular carcinoma (HCC) in all treated MMA mice and control littermates. AAV-mediated genome editing of MMUT into the albumin locus resulted in permanent hepatic correction in MMA mouse models, which was accompanied by decreased levels of methylmalonate and Fgf21, and improved survival without HCC. With time, levels of transgene expression increased and methylmalonate progressively decreased, whereas the number of albumin-MMUT integrations and corrected hepatocytes in MMA mice increased, but not in similarly treated wild-type animals. Additionally, expression of MMUT in the setting of MMA conferred a selective growth advantage upon edited cells, which potentiates the therapeutic response. CONCLUSIONS: In conclusion, our findings demonstrate that AAV-mediated, promoterless, nuclease-free genome editing at the albumin locus provides safe and durable therapeutic benefit in neonatally treated MMA mice.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/terapia , Dependovirus/genética , Edição de Genes/métodos , Terapia Genética/métodos , Metilmalonil-CoA Mutase/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/sangue , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/sangue , Hepatócitos , Neoplasias Hepáticas/patologia , Transplante de Fígado , Malonatos/sangue , Metilmalonil-CoA Mutase/genética , Camundongos , Camundongos Endogâmicos C57BL
9.
Nat Commun ; 11(1): 5851, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33203857

RESUMO

HIV viremia can be controlled by chronic antiretroviral therapy. As a potentially single-shot alternative, B cells engineered by CRISPR/Cas9 to express anti-HIV broadly neutralizing antibodies (bNAbs) are capable of secreting high antibody titers. Here, we show that, upon immunization of mice, adoptively transferred engineered B cells home to germinal centers (GC) where they predominate over the endogenous response and differentiate into memory and plasma cells while undergoing class switch recombination (CSR). Immunization with a high affinity antigen increases accumulation in GCs and CSR rates. Boost immunization increases the rate of engineered B cells in GCs and antibody secretion, indicating memory retention. Finally, antibody sequences of engineered B cells in the spleen show patterns of clonal selection. Therefore, B cells can be engineered into what could be a living and evolving drug.


Assuntos
Vacinas contra a AIDS/imunologia , Anticorpos Monoclonais/genética , Linfócitos B/imunologia , Anticorpos Amplamente Neutralizantes/genética , Anticorpos Anti-HIV/genética , Memória Imunológica/genética , Vacinas contra a AIDS/genética , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Linfócitos B/fisiologia , Linfócitos B/transplante , Anticorpos Amplamente Neutralizantes/sangue , Anticorpos Amplamente Neutralizantes/imunologia , Engenharia Genética/métodos , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , Imunização , Isotipos de Imunoglobulinas/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação
10.
JCI Insight ; 52019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31211694

RESUMO

Non-integrative AAV-mediated gene therapy in the liver is effective in adult patients, but faces limitations in pediatric settings due to episomal DNA loss during hepatocyte proliferation. Gene targeting is a promising approach by permanently modifying the genome. We previously rescued neonatal lethality in Crigler-Najjar mice by inserting a promoterless human uridine glucuronosyl transferase A1 (UGT1A1) cDNA in exon 14 of the albumin gene, without the use of nucleases. To increase recombination rate and therapeutic efficacy, here we used CRISPR/SaCas9. Neonatal mice were transduced with two AAVs: one expressing the SaCas9 and sgRNA, and one containing a promoterless cDNA flanked by albumin homology regions. Targeting efficiency increased ~26-fold with an eGFP reporter cDNA, reaching up to 24% of eGFP-positive hepatocytes. Next, we fully corrected the diseased phenotype of Crigler-Najjar mice by targeting the hUGT1A1 cDNA. Treated mice had normal plasma bilirubin up to 10 months after administration, hUGT1A1 protein levels were ~6-fold higher than in WT liver, with a 90-fold increase in recombination rate. Liver histology, inflammatory markers, and plasma albumin were normal in treated mice, with no off-targets in predicted sites. Thus, the improved efficacy and reassuring safety profile support the potential application of the proposed approach to other liver diseases.


Assuntos
Marcação de Genes/métodos , Terapia Genética/métodos , Glucuronosiltransferase/genética , Fígado/metabolismo , Doenças Metabólicas/genética , Doenças Metabólicas/terapia , Animais , Animais Recém-Nascidos , Bilirrubina , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA Complementar , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Glucuronosiltransferase/metabolismo , Células HEK293 , Hepatócitos/metabolismo , Humanos , Fígado/patologia , Masculino , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Albumina Sérica , Usos Terapêuticos
13.
Mol Ther ; 25(11): 2477-2489, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29032169

RESUMO

Hepatocytes represent an important target for gene therapy and editing of single-gene disorders. In α-1 antitrypsin (AAT) deficiency, one missense mutation results in impaired secretion of AAT. In most patients, lung damage occurs due to a lack of AAT-mediated protection of lung elastin from neutrophil elastase. In some patients, accumulation of misfolded PiZ mutant AAT protein triggers hepatocyte injury, leading to inflammation and cirrhosis. We hypothesized that correcting the Z mutant defect in hepatocytes would confer a selective advantage for repopulation of hepatocytes within an intact liver. A human PiZ allele was crossed onto an immune-deficient (NSG) strain to create a recipient strain (NSG-PiZ) for human hepatocyte xenotransplantation. Results indicate that NSG-PiZ recipients support heightened engraftment of normal human primary hepatocytes as compared with NSG recipients. This model can therefore be used to test hepatocyte cell therapies for AATD, but more broadly it serves as a simple, highly reproducible liver xenograft model. Finally, a promoterless adeno-associated virus (AAV) vector, expressing a wild-type AAT and a synthetic miRNA to silence the endogenous allele, was integrated into the albumin locus. This gene-editing approach leads to a selective advantage of edited hepatocytes, by silencing the mutant protein and augmenting normal AAT production, and improvement of the liver pathology.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Hepatócitos/transplante , Transgenes , Deficiência de alfa 1-Antitripsina/terapia , alfa 1-Antitripsina/genética , Animais , Dependovirus/metabolismo , Modelos Animais de Doenças , Edição de Genes , Expressão Gênica , Inativação Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Sobrevivência de Enxerto , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação , Transplante Heterólogo , alfa 1-Antitripsina/metabolismo , Deficiência de alfa 1-Antitripsina/enzimologia , Deficiência de alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/patologia
14.
EMBO Mol Med ; 9(10): 1346-1355, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28751579

RESUMO

Crigler-Najjar syndrome type I (CNSI) is a rare monogenic disease characterized by severe neonatal unconjugated hyperbilirubinemia with a lifelong risk of neurological damage and death. Liver transplantation is the only curative option, which has several limitations and risks. We applied an in vivo gene targeting approach based on the insertion, without the use of nucleases, of a promoterless therapeutic cDNA into the albumin locus of a mouse model reproducing all major features of CNSI Neonatal transduction with the donor vector resulted in the complete rescue from neonatal lethality, with a therapeutic reduction in plasma bilirubin lasting for at least 12 months, the latest time point analyzed. Mutant mice, which expressed about 5-6% of WT Ugt1a1 levels, showed normal liver histology and motor-coordination abilities, suggesting no functional liver or brain abnormalities. These results proved that the promoterless gene therapy is applicable for CNSI, providing therapeutic levels of an intracellular ER membrane-bound enzyme responsible for a lethal liver metabolic disease.


Assuntos
Síndrome de Crigler-Najjar/terapia , Marcação de Genes/métodos , Terapia Genética/métodos , Regiões Promotoras Genéticas , Animais , Bilirrubina/sangue , Encéfalo/patologia , Síndrome de Crigler-Najjar/genética , Modelos Animais de Doenças , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/terapia , Fígado/patologia , Camundongos , Camundongos Mutantes , Análise de Sobrevida , Transdução Genética
15.
Sci Transl Med ; 8(342): 342ra79, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27280686

RESUMO

Many genetic and acquired liver disorders are amenable to gene and/or cell therapy. However, the efficiencies of cell engraftment and stable genetic modification are low and often subtherapeutic. In particular, targeted gene modifications from homologous recombination are rare events. These obstacles could be overcome if hepatocytes that have undergone genetic modification were to be selectively amplified or expanded. We describe a universally applicable system for in vivo selection and expansion of gene-modified hepatocytes in any genetic background. In this system, the therapeutic transgene is coexpressed with a short hairpin RNA (shRNA) that confers modified hepatocytes with resistance to drug-induced toxicity. An shRNA against the tyrosine catabolic enzyme 4-OH-phenylpyruvate dioxygenase protected hepatocytes from 4-[(2-carboxyethyl)-hydroxyphosphinyl]-3-oxobutyrate, a small-molecule inhibitor of fumarylacetoacetate hydrolase. To select for specific gene targeting events, the protective shRNA was embedded in a microRNA and inserted into a recombinant adeno-associated viral vector designed to integrate site-specifically into the highly active albumin locus. After selection of the gene-targeted cells, transgene expression increased 10- to 1000-fold, reaching supraphysiological levels of human factor 9 protein (50,000 ng/ml) in mice. This drug resistance system can be used to achieve therapeutically relevant transgene levels in hepatocytes in any setting.


Assuntos
Hepatócitos/metabolismo , Animais , Dependovirus/genética , Hidrolases/antagonistas & inibidores , Hidrolases/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Mutantes , MicroRNAs/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
16.
BMC Evol Biol ; 11: 324, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-22054298

RESUMO

BACKGROUND: Homing endonucleases (HEases) are a large and diverse group of site-specific DNAases. They reside within self-splicing introns and inteins, and promote their horizontal dissemination. In recent years, HEases have been the focus of extensive research due to their promising potential use in gene targeting procedures for the treatment of genetic diseases and for the genetic engineering of crop, animal models and cell lines. RESULTS: Using mathematical analysis and computational modeling, we present here a novel account for the evolution and population dynamics of HEase genes (HEGs). We describe HEGs as paradoxical selfish elements whose long-term persistence in a single population relies on low transmission rates and a positive correlation between transmission efficiency and toxicity. CONCLUSION: Plausible conditions allow HEGs to sustain at high frequency through long evolutionary periods, with the endonuclease frequency being either at equilibrium or periodically oscillating. The predictions of our model may prove important not only for evolutionary theory but also for gene therapy and bio-engineering applications of HEases.


Assuntos
Endonucleases/genética , Evolução Molecular , Alelos , Animais , Simulação por Computador , Humanos , Íntrons , Modelos Genéticos
17.
Nucleic Acids Res ; 39(15): 6646-59, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21525128

RESUMO

In recent years, both homing endonucleases (HEases) and zinc-finger nucleases (ZFNs) have been engineered and selected for the targeting of desired human loci for gene therapy. However, enzyme engineering is lengthy and expensive and the off-target effect of the manufactured endonucleases is difficult to predict. Moreover, enzymes selected to cleave a human DNA locus may not cleave the homologous locus in the genome of animal models because of sequence divergence, thus hampering attempts to assess the in vivo efficacy and safety of any engineered enzyme prior to its application in human trials. Here, we show that naturally occurring HEases can be found, that cleave desirable human targets. Some of these enzymes are also shown to cleave the homologous sequence in the genome of animal models. In addition, the distribution of off-target effects may be more predictable for native HEases. Based on our experimental observations, we present the HomeBase algorithm, database and web server that allow a high-throughput computational search and assignment of HEases for the targeting of specific loci in the human and other genomes. We validate experimentally the predicted target specificity of candidate fungal, bacterial and archaeal HEases using cell free, yeast and archaeal assays.


Assuntos
Endodesoxirribonucleases/metabolismo , Marcação de Genes , Algoritmos , Animais , Archaea/enzimologia , Bactérias/enzimologia , Sequência de Bases , Sequência Conservada , Bases de Dados de Ácidos Nucleicos , Endodesoxirribonucleases/genética , Fungos/enzimologia , Humanos , Modelos Animais , Mutação
18.
PLoS One ; 6(1): e15833, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21283796

RESUMO

Inteins are parasitic genetic elements, analogous to introns that excise themselves at the protein level by self-splicing, allowing the formation of functional non-disrupted proteins. Many inteins contain a homing endonuclease (HEN) gene, and rely on its activity for horizontal propagation. In the halophilic archaeon, Haloferax volcanii, the gene encoding DNA polymerase B (polB) contains an intein with an annotated but uncharacterized HEN. Here we examine the activity of the polB HEN in vivo, within its natural archaeal host. We show that this HEN is highly active, and able to insert the intein into both a chromosomal target and an extra-chromosomal plasmid target, by gene conversion. We also demonstrate that the frequency of its incorporation depends on the length of the flanking homologous sequences around the target site, reflecting its dependence on the homologous recombination machinery. Although several evolutionary models predict that the presence of an intein involves a change in the fitness of the host organism, our results show that a strain deleted for the intein sequence shows no significant changes in growth rate compared to the wild type.


Assuntos
DNA Polimerase Dirigida por DNA/genética , Endonucleases/genética , Haloferax volcanii/enzimologia , Evolução Biológica , Cromossomos/genética , Conversão Gênica , Aptidão Genética , Inteínas/genética , Plasmídeos/genética
19.
Biochem Soc Trans ; 39(1): 169-73, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21265767

RESUMO

Inteins are selfish genetic elements that disrupt the sequence of protein-coding genes and are excised post-translationally. Most inteins also contain a HEN (homing endonuclease) domain, which is important for their horizontal transmission. The present review focuses on the evolution of inteins and their nested HENs, and highlights several unsolved questions that could benefit from molecular genetic approaches. Such approaches can be well carried out in halophilic archaea, which are naturally intein-rich and have highly developed genetic tools for their study. In particular, the fitness effects of harbouring an intein/HEN can be tested in direct competition assays, providing additional insights that will improve current evolutionary models.


Assuntos
Endonucleases/química , Endonucleases/genética , Evolução Molecular , Inteínas/genética , Archaea/genética , Archaea/metabolismo , Endonucleases/metabolismo , Mutação
20.
Nat Rev Genet ; 9(1): 27-37, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18040271

RESUMO

Decades of research into homologous recombination have unravelled many of the details concerning the transfer of information between two homologous sequences. By contrast, the processes by which the interacting molecules initially colocalize are largely unknown. How can two homologous needles find each other in the genomic haystack? Is homologous pairing the result of a damage-induced homology search, or is it an enduring and general feature of the genomic architecture that facilitates homologous recombination whenever and wherever damage occurs? This Review presents the homologous-pairing enigma, delineates our current understanding of the process and offers guidelines for future research.


Assuntos
Recombinação Genética , Homologia de Sequência do Ácido Nucleico , Dano ao DNA , Reparo do DNA , Saccharomyces cerevisiae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...