Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 383(6689): 1344-1349, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38513017

RESUMO

Large DNA assembly methodologies underlie milestone achievements in synthetic prokaryotic and budding yeast chromosomes. While budding yeast control chromosome inheritance through ~125-base pair DNA sequence-defined centromeres, mammals and many other eukaryotes use large, epigenetic centromeres. Harnessing centromere epigenetics permits human artificial chromosome (HAC) formation but is not sufficient to avoid rampant multimerization of the initial DNA molecule upon introduction to cells. We describe an approach that efficiently forms single-copy HACs. It employs a ~750-kilobase construct that is sufficiently large to house the distinct chromatin types present at the inner and outer centromere, obviating the need to multimerize. Delivery to mammalian cells is streamlined by employing yeast spheroplast fusion. These developments permit faithful chromosome engineering in the context of metazoan cells.


Assuntos
Centrômero , Cromossomos Artificiais Humanos , Epigênese Genética , Humanos , Centrômero/genética , Centrômero/metabolismo , Cromatina/metabolismo , Cromossomos Artificiais Humanos/genética , Cromossomos Artificiais Humanos/metabolismo , Saccharomycetales/genética
2.
Chromosome Res ; 31(3): 17, 2023 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-37353691

RESUMO

Telomerase is a ribonucleoprotein ribonucleic enzyme that elongates telomere repeat sequences at the ends of chromosomes and contributes to cellular immortalization. The catalytic component of telomerase, human telomerase reverse transcriptase (hTERT), has been observed to be reactivated in immortalized cells. Notably, most cancer cells have been found to have active hTERT mRNA transcription, resulting in continuous cell division, which is crucial for malignant transformation. Therefore, discovering mechanisms underlying the regulation of hTERT transcription is an attractive target for cancer-specific treatments.Loss of heterozygosity (LOH) of chromosome 3p21.3 has been frequently observed in human oral squamous cell carcinoma (OSCC). Moreover, we previously reported that HSC3 OSCC microcell hybrid clones with an introduced human chromosome 3 (HSC3#3) showed inhibition of hTERT transcription compared with the parental HSC3 cells. This study examined whether hTERT transcription regulators are present in the 3p21.3 region. We constructed a human artificial chromosome (HAC) vector (3p21.3-HAC) with only the 3p21.3-p22.2 region and performed functional analysis using the 3p21.3-HAC. HSC3 microcell hybrid clones with an introduced 3p21.3-HAC exhibited significant suppression of hTERT transcription, similar to the microcell hybrid clones with an intact chromosome 3. In contrast, HSC3 clones with truncated chromosome 3 with deletion of the 3p21.3 region (3delp21.3) showed no effect on hTERT expression levels. These results provide direct evidence that hTERT suppressor gene(s) were retained in the 3p21.3 region, suggesting that the presence of regulatory factors that control telomerase enzyme activity may be involved in the development of OSCC.


Assuntos
Carcinoma de Células Escamosas , Cromossomos Artificiais Humanos , Neoplasias Bucais , Telomerase , Humanos , Telomerase/genética , Telomerase/metabolismo , Carcinoma de Células Escamosas/genética , Cromossomos Artificiais Humanos/metabolismo , Neoplasias Bucais/genética , Transcrição Gênica
3.
ACS Synth Biol ; 9(12): 3267-3287, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33289546

RESUMO

Human artificial chromosomes (HACs) are important tools for epigenetic engineering, for measuring chromosome instability (CIN), and for possible gene therapy. However, their use in the latter is potentially limited because the input HAC-seeding DNA can undergo an unpredictable series of rearrangements during HAC formation. As a result, after transfection and HAC formation, each cell clone contains a HAC with a unique structure that cannot be precisely predicted from the structure of the HAC-seeding DNA. Although it has been reported that these rearrangements can happen, the timing and mechanism of their formation has yet to be described. Here we synthesized a HAC-seeding DNA with two distinct structural domains and introduced it into HT1080 cells. We characterized a number of HAC-containing clones and subclones to track DNA rearrangements during HAC establishment. We demonstrated that rearrangements can occur early during HAC formation. Subsequently, the established HAC genomic organization is stably maintained across many cell generations. Thus, early stages in HAC formation appear to at least occasionally involve a process of DNA shredding and shuffling that resembles chromothripsis, an important hallmark of many cancer types. Understanding these events during HAC formation has critical implications for future efforts aimed at synthesizing and exploiting synthetic human chromosomes.


Assuntos
Cromossomos Artificiais Humanos/metabolismo , Rearranjo Gênico/fisiologia , Linhagem Celular Tumoral , Centrômero/metabolismo , Proteína B de Centrômero/genética , Instabilidade Cromossômica , Epigênese Genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos
4.
J Cell Sci ; 133(14)2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32576667

RESUMO

Most eukaryotic centromeres are located within heterochromatic regions. Paradoxically, heterochromatin can also antagonize de novo centromere formation, and some centromeres lack it altogether. In order to investigate the importance of heterochromatin at centromeres, we used epigenetic engineering of a synthetic alphoidtetO human artificial chromosome (HAC), to which chimeric proteins can be targeted. By tethering the JMJD2D demethylase (also known as KDM4D), we removed heterochromatin mark H3K9me3 (histone 3 lysine 9 trimethylation) specifically from the HAC centromere. This caused no short-term defects, but long-term tethering reduced HAC centromere protein levels and triggered HAC mis-segregation. However, centromeric CENP-A was maintained at a reduced level. Furthermore, HAC centromere function was compatible with an alternative low-H3K9me3, high-H3K27me3 chromatin signature, as long as residual levels of H3K9me3 remained. When JMJD2D was released from the HAC, H3K9me3 levels recovered over several days back to initial levels along with CENP-A and CENP-C centromere levels, and mitotic segregation fidelity. Our results suggest that a minimal level of heterochromatin is required to stabilize mitotic centromere function but not for maintaining centromere epigenetic memory, and that a homeostatic pathway maintains heterochromatin at centromeres.This article has an associated First Person interview with the first authors of the paper.


Assuntos
Cromossomos Artificiais Humanos , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Segregação de Cromossomos/genética , Cromossomos Artificiais Humanos/genética , Cromossomos Artificiais Humanos/metabolismo , Epigênese Genética , Heterocromatina , Histonas/genética , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji , Cinetocoros/metabolismo
5.
Nucleic Acids Res ; 48(1): 486-499, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31745563

RESUMO

Cross-species pathway transplantation enables insight into a biological process not possible through traditional approaches. We replaced the enzymes catalyzing the entire Saccharomyces cerevisiae adenine de novo biosynthesis pathway with the human pathway. While the 'humanized' yeast grew in the absence of adenine, it did so poorly. Dissection of the phenotype revealed that PPAT, the human ortholog of ADE4, showed only partial function whereas all other genes complemented fully. Suppressor analysis revealed other pathways that play a role in adenine de-novo pathway regulation. Phylogenetic analysis pointed to adaptations of enzyme regulation to endogenous metabolite level 'setpoints' in diverse organisms. Using DNA shuffling, we isolated specific amino acids combinations that stabilize the human protein in yeast. Thus, using adenine de novo biosynthesis as a proof of concept, we suggest that the engineering methods used in this study as well as the debugging strategies can be utilized to transplant metabolic pathway from any origin into yeast.


Assuntos
Adenina/biossíntese , Vias Biossintéticas/genética , Carboxiliases/genética , Cromossomos Artificiais Humanos/química , Peptídeo Sintases/genética , Saccharomyces cerevisiae/genética , Sequência de Aminoácidos , Sistemas CRISPR-Cas , Carboxiliases/metabolismo , Cromossomos Artificiais Humanos/metabolismo , Teste de Complementação Genética , Engenharia Genética/métodos , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Peptídeo Sintases/metabolismo , Filogenia , Plasmídeos/química , Plasmídeos/metabolismo , Saccharomyces cerevisiae/classificação , Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
6.
Cell ; 178(3): 624-639.e19, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31348889

RESUMO

Recent breakthroughs with synthetic budding yeast chromosomes expedite the creation of synthetic mammalian chromosomes and genomes. Mammals, unlike budding yeast, depend on the histone H3 variant, CENP-A, to epigenetically specify the location of the centromere-the locus essential for chromosome segregation. Prior human artificial chromosomes (HACs) required large arrays of centromeric α-satellite repeats harboring binding sites for the DNA sequence-specific binding protein, CENP-B. We report the development of a type of HAC that functions independently of these constraints. Formed by an initial CENP-A nucleosome seeding strategy, a construct lacking repetitive centromeric DNA formed several self-sufficient HACs that showed no uptake of genomic DNA. In contrast to traditional α-satellite HAC formation, the non-repetitive construct can form functional HACs without CENP-B or initial CENP-A nucleosome seeding, revealing distinct paths to centromere formation for different DNA sequence types. Our developments streamline the construction and characterization of HACs to facilitate mammalian synthetic genome efforts.


Assuntos
Centrômero/metabolismo , Cromossomos Artificiais Humanos/metabolismo , DNA Satélite/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Centrômero/genética , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Proteína B de Centrômero/deficiência , Proteína B de Centrômero/genética , Proteína B de Centrômero/metabolismo , Epigênese Genética , Humanos , Nucleossomos/química , Nucleossomos/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo
7.
ACS Synth Biol ; 7(9): 1974-1989, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30075081

RESUMO

Since their description in the late 1990s, Human Artificial Chromosomes (HACs) bearing functional kinetochores have been considered as promising systems for gene delivery and expression. More recently a HAC assembled from a synthetic alphoid DNA array has been exploited in studies of centromeric chromatin and in assessing the impact of different epigenetic modifications on kinetochore structure and function in human cells. This HAC was termed the alphoidtetO-HAC, as the synthetic monomers each contained a tetO sequence in place of the CENP-B box that can be targeted specifically with tetR-fusion proteins. Studies in which the kinetochore chromatin of the alphoidtetO-HAC was specifically modified, revealed that heterochromatin is incompatible with centromere function and that centromeric transcription is important for centromere assembly and maintenance. In addition, the alphoidtetO-HAC was modified to carry large gene inserts that are expressed in target cells under conditions that recapitulate the physiological regulation of endogenous loci. Importantly, the phenotypes arising from stable gene expression can be reversed when cells are "cured" of the HAC by inactivating its kinetochore in proliferating cell populations, a feature that provides a control for phenotypic changes attributed to expression of HAC-encoded genes. AlphoidtetO-HAC-based technology has also been used to develop new drug screening and assessment strategies to manipulate the CIN phenotype in cancer cells. In summary, the alphoidtetO-HAC is proving to be a versatile tool for studying human chromosome transactions and structure as well as for genome and cancer studies.


Assuntos
Centrômero/metabolismo , Cromossomos Artificiais Humanos/genética , Neoplasias/patologia , Animais , Proteína B de Centrômero/genética , Proteína B de Centrômero/metabolismo , Instabilidade Cromossômica , Cromossomos Artificiais Humanos/metabolismo , Técnicas de Transferência de Genes , Histonas/metabolismo , Humanos , Neoplasias/genética
8.
Drug Metab Pharmacokinet ; 33(1): 17-30, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29398301

RESUMO

In the earliest stage of drug discovery/development, various cell-based models and animal models were used for the prediction of human pharmacokinetics and toxicokinetics. Unfortunately, drugs under development are often discontinued because their nonclinical results do not extrapolate to human clinical studies in relation to either safety or efficacy. Therefore, it is important to improve the time- and cost-effectiveness of drug development. This might be achieved by developing new technologies including pharmacokinetics and toxicokinetics models that use human and mouse artificial chromosome vectors (HACs/MACs). HACs/MACs are unique vectors with several advantages: 1) independent maintenance, 2) defined copy number and mitotically stable, 3) no silencing of the transgene, and 4) no limitation of DNA insertion size. This review provides information on the advantages and examples of the utility of various models based on the recent advances in HAC/MAC technologies, including multifunctional cell-based models for assaying drug-drug interactions, bidirectional permeability, and cytotoxicity, as well as fully genetically humanized mouse models. We also discuss the future prospects of these technologies to advance drug discovery. In summary, these technologies offer advantages over current conventional models and should improve the success rate of drug development related to efficacy and safety for humans.


Assuntos
Cromossomos Artificiais/metabolismo , Descoberta de Drogas/métodos , Técnicas de Transferência de Genes , Vetores Genéticos/farmacocinética , Animais , Cromossomos Artificiais/genética , Cromossomos Artificiais Humanos/genética , Cromossomos Artificiais Humanos/metabolismo , Clonagem Molecular/métodos , Descoberta de Drogas/tendências , Técnicas de Transferência de Genes/tendências , Vetores Genéticos/genética , Humanos , Camundongos , Toxicocinética
9.
Genome Res ; 27(6): 922-933, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28341771

RESUMO

The spatial arrangement of chromatin is linked to the regulation of nuclear processes. One striking aspect of nuclear organization is the spatial segregation of heterochromatic and euchromatic domains. The mechanisms of this chromatin segregation are still poorly understood. In this work, we investigated the link between the primary genomic sequence and chromatin domains. We analyzed the spatial intranuclear arrangement of a human artificial chromosome (HAC) in a xenospecific mouse background in comparison to an orthologous region of native mouse chromosome. The two orthologous regions include segments that can be assigned to three major chromatin classes according to their gene abundance and repeat repertoire: (1) gene-rich and SINE-rich euchromatin; (2) gene-poor and LINE/LTR-rich heterochromatin; and (3) gene-depleted and satellite DNA-containing constitutive heterochromatin. We show, using fluorescence in situ hybridization (FISH) and 4C-seq technologies, that chromatin segments ranging from 0.6 to 3 Mb cluster with segments of the same chromatin class. As a consequence, the chromatin segments acquire corresponding positions in the nucleus irrespective of their chromosomal context, thereby strongly suggesting that this is their autonomous property. Interactions with the nuclear lamina, although largely retained in the HAC, reveal less autonomy. Taken together, our results suggest that building of a functional nucleus is largely a self-organizing process based on mutual recognition of chromosome segments belonging to the major chromatin classes.


Assuntos
Núcleo Celular/genética , Cromossomos Artificiais Humanos/metabolismo , Eucromatina/metabolismo , Fibroblastos/metabolismo , Heterocromatina/metabolismo , Retina/metabolismo , Animais , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Cromossomos Artificiais Humanos/ultraestrutura , Eucromatina/classificação , Eucromatina/ultraestrutura , Fibroblastos/ultraestrutura , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Heterocromatina/classificação , Heterocromatina/ultraestrutura , Humanos , Hibridização in Situ Fluorescente , Camundongos , Cultura Primária de Células , Retina/ultraestrutura
10.
Iran Biomed J ; 20(2): 68-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26602035

RESUMO

Mammalian expression systems, due to their capacity in post-translational modification, are preferred systems for biopharmaceutical protein production. Several recombinant protein systems have been introduced to the market, most of which are under clinical development. In spite of significant improvements such as cell line engineering, introducing novel expression methods, gene silencing and process development, expression level is unpredictable and unstable because of the random location of integration in the genome. Site-specific recombination techniques are capable of producing stable and high producer clonal cells; therefore, they are gaining more importance in the biopharmaceutical production. Site-specific recombination methods increase the recombinant protein production by specifically inserting a vector at a locus with specific expression trait. The present review focused on the latest developments in site-specific recombination techniques, their specific features and comparisons.


Assuntos
Biofarmácia/métodos , Engenharia Genética/métodos , Proteínas Recombinantes/genética , Recombinação Genética/genética , Animais , Cromossomos Artificiais Humanos/genética , Cromossomos Artificiais Humanos/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Humanos , Proteínas Recombinantes/metabolismo
12.
Cell Cycle ; 14(8): 1268-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25695642

RESUMO

De novo assembled alphoid(tetO)-type human artificial chromosomes (HACs) represent a novel promising generation of high capacity episomal vectors. Their function and persistence, and any adverse effects, in various cell types in live animals, have not, however, been explored. In this study we transferred the alphoid(tetO)-HAC into mouse ES cells and assessed whether the presence of this extra chromosome affects their pluripotent properties. Alphoid(tetO)-HAC-bearing ES cells were indistinguishable from their wild-type counterparts: they retained self-renewal potential and full capacity for multilineage differentiation during mouse development, whereas the HAC itself was mitotically and transcriptionally stable during this process. Our data provide the first example of fully synthetic DNA behaving like a normal chromosome in cells of living animals. It also opens a new perspective into functional genetic studies in laboratory animals as well as stem cell-based regenerative medicine.


Assuntos
Diferenciação Celular , Cromossomos Artificiais Humanos/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Células CHO , Cromossomos Artificiais Humanos/genética , Cricetinae , Cricetulus , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Teratoma/metabolismo , Teratoma/patologia
13.
Methods Mol Biol ; 1227: 3-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25239739

RESUMO

Transformation-associated recombination (TAR) cloning allows selective isolation of full-length genes and genomic loci as large circular Yeast Artificial Chromosomes (YACs) in yeast. The method has a broad application for structural and functional genomics, long-range haplotyping, characterization of chromosomal rearrangements, and evolutionary studies. In this paper, we describe a basic protocol for gene isolation by TAR as well as a method to convert TAR isolates into Bacterial Artificial Chromosomes (BACs) using a retrofitting vector. The retrofitting vector contains a 3' HPRT-loxP cassette to allow subsequent gene loading into a unique loxP site of the HAC-based (Human Artificial Chromosome) gene delivery vector. The benefit of combining the TAR gene cloning technology with the HAC gene delivery system for gene expression studies is discussed.


Assuntos
Clonagem Molecular/métodos , DNA Fúngico/genética , Regulação Fúngica da Expressão Gênica , Saccharomyces cerevisiae/genética , Esferoplastos/genética , Animais , Células CHO , Cromossomos Artificiais Bacterianos/química , Cromossomos Artificiais Bacterianos/metabolismo , Cromossomos Artificiais Humanos/química , Cromossomos Artificiais Humanos/metabolismo , Cromossomos Artificiais de Levedura/química , Cromossomos Artificiais de Levedura/metabolismo , Cricetulus , DNA Fúngico/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Recombinação Genética , Saccharomyces cerevisiae/metabolismo , Esferoplastos/metabolismo , Transformação Genética
14.
PLoS One ; 8(10): e78119, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205120

RESUMO

Therapeutic human polyclonal antibodies (hpAbs) derived from pooled plasma from human donors are Food and Drug Administration approved biologics used in the treatment of a variety of human diseases. Powered by the natural diversity of immune response, hpAbs are effective in treating diseases caused by complex or quickly-evolving antigens such as viruses. We previously showed that transchromosomic (Tc) cattle carrying a human artificial chromosome (HAC) comprising the entire unrearranged human immunoglobulin heavy-chain (hIGH) and kappa-chain (hIGK) germline loci (named as κHAC) are capable of producing functional hpAbs when both of the bovine immunoglobulin mu heavy-chains, bIGHM and bIGHML1, are homozygously inactivated (double knockouts or DKO). However, B lymphocyte development in these Tc cattle is compromised, and the overall production of hpAbs is low. Here, we report the construction of an improved HAC, designated as cKSL-HACΔ, by incorporating all of the human immunoglobulin germline loci into the HAC. Furthermore, for avoiding the possible human-bovine interspecies incompatibility between the human immunoglobulin mu chain protein (hIgM) and bovine transmembrane α and ß immunoglobulins (bIgα and bIgß) in the pre-B cell receptor (pre-BCR) complex, we partially replaced (bovinized) the hIgM constant domain with the counterpart of bovine IgM (bIgM) that is involved in the interaction between bIgM and bIgα/Igß; human IgM bovinization would also improve the functionality of hIgM in supporting B cell activation and proliferation. We also report the successful production of DKO Tc cattle carrying the cKSL-HACΔ (cKSL-HACΔ/DKO), the dramatic improvement of B cell development in these cattle and the high level production of hpAbs (as measured for the human IgG isotype) in the plasma. We further demonstrate that, upon immunization by tumor immunogens, high titer tumor immunogen-specific human IgG (hIgG) can be produced from such Tc cattle.


Assuntos
Imunoglobulinas/metabolismo , Animais , Linfócitos B/metabolismo , Bovinos , Proliferação de Células , Cromossomos Artificiais Humanos/genética , Cromossomos Artificiais Humanos/imunologia , Cromossomos Artificiais Humanos/metabolismo , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Cadeias mu de Imunoglobulina/genética , Cadeias mu de Imunoglobulina/imunologia , Cadeias mu de Imunoglobulina/metabolismo , Imunoglobulinas/genética , Imunoglobulinas/imunologia
15.
Eur J Hum Genet ; 21(9): 948-56, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23403904

RESUMO

Human artificial chromosomes (HAC) are a valuable tool in the analysis of complex chromatin structures such as the human centromere because of their small size and relative simplicity compared with normal human chromosomes. This report includes a comprehensive study of the centromere and chromatin composition of HAC, expressing human genes, generated in human cells and transferred to murine cells. The analysis involved chromatin immuno-precipitation and immuno-FISH on metaphase chromosomes and chromatin fibres. In both the cell types, the HAC consisted of alphoid and non-alphoid DNA and were mainly euchromatic in composition, although a pericentromeric heterochromatic region was present on all the HAC. Fibre-FISH and chromatin immuno-precipitation data indicated that the position of the centromere differed between HAC in human cells and in murine cells. Our work highlights the importance and utilisation of HAC for understanding the epigenetic aspects of chromosome biology.


Assuntos
Centrômero/genética , Cromossomos Artificiais Humanos/genética , Animais , Linhagem Celular , Centrômero/metabolismo , Instabilidade Cromossômica , Cromossomos Artificiais Humanos/metabolismo , Humanos , Hibridização in Situ Fluorescente , Metáfase , Camundongos , Conformação de Ácido Nucleico
16.
Chromosome Res ; 20(5): 505-19, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22825423

RESUMO

The centromere is a specialized chromosomal region that serves as the assembly site of the kinetochore. At the centromere, CENP-A nucleosomes form part of a chromatin landscape termed centrochromatin. This chromatin environment conveys epigenetic marks regulating kinetochore formation. Recent work sheds light on the intricate relationship between centrochromatin state, the CENP-A assembly pathway and the maintenance of centromere function. Here, we review the emerging picture of how chromatin affects mammalian kinetochore formation. We place particular emphasis on data obtained from Human Artificial Chromosome (HAC) biology and the targeted engineering of centrochromatin using synthetic HACs. We discuss implications of these findings, which indicate that a delicate balance of histone modifications and chromatin state dictates both de novo centromere formation and the maintenance of centromere identity in dividing cell populations.


Assuntos
Autoantígenos/metabolismo , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos Artificiais Humanos/metabolismo , Proteína Centromérica A , Proteína B de Centrômero/metabolismo , Montagem e Desmontagem da Cromatina , DNA Satélite/metabolismo , Vetores Genéticos , Heterocromatina/metabolismo , Humanos , Mitose , Nucleossomos/metabolismo , Transcrição Gênica
17.
Sci Transl Med ; 3(96): 96ra78, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21849666

RESUMO

In contrast to conventional gene therapy vectors, human artificial chromosomes (HACs) are episomal vectors that can carry large regions of the genome containing regulatory elements. So far, HACs have not been used as vectors in gene therapy for treating genetic disorders. Here, we report the amelioration of the dystrophic phenotype in the mdx mouse model of Duchenne muscular dystrophy (DMD) using a combination of HAC-mediated gene replacement and transplantation with blood vessel-associated stem cells (mesoangioblasts). We first genetically corrected mesoangioblasts from dystrophic mdx mice with a HAC vector containing the entire (2.4 Mb) human dystrophin genetic locus. Genetically corrected mesoangioblasts engrafted robustly and gave rise to many dystrophin-positive muscle fibers and muscle satellite cells in dystrophic mice, leading to morphological and functional amelioration of the phenotype that lasted for up to 8 months after transplantation. Thus, HAC-mediated gene transfer shows efficacy in a preclinical model of DMD and offers potential for future clinical translation.


Assuntos
Cromossomos Artificiais Humanos/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Células-Tronco/citologia , Animais , Cromossomos Artificiais Humanos/genética , Distrofina/genética , Distrofina/metabolismo , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/genética , Transplante de Células-Tronco , Células-Tronco/metabolismo
18.
Hum Mol Genet ; 20(15): 2905-13, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21593218

RESUMO

We present a novel and efficient non-integrating gene expression system in human embryonic stem cells (hESc) utilizing human artificial chromosomes (HAC), which behave as autonomous endogenous host chromosomes and segregate correctly during cell division. HAC are important vectors for investigating the organization and structure of the kinetochore, and gene complementation. HAC have so far been obtained in immortalized or tumour-derived cell lines, but never in stem cells, thus limiting their potential therapeutic application. In this work, we modified the herpes simplex virus type 1 amplicon system for efficient transfer of HAC DNA into two hESc. The deriving stable clones generated green fluorescent protein gene-expressing HAC at high frequency, which were stably maintained without selection for 3 months. Importantly, no integration of the HAC DNA was observed in the hESc lines, compared with the fibrosarcoma-derived control cells, where the exogenous DNA frequently integrated in the host genome. The hESc retained pluripotency, differentiation and teratoma formation capabilities. This is the first report of successfully generating gene expressing de novo HAC in hESc, and is a significant step towards the genetic manipulation of stem cells and potential therapeutic applications.


Assuntos
Cromossomos Artificiais Humanos/metabolismo , Células-Tronco Embrionárias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Cromossomos Artificiais Humanos/genética , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Imunofluorescência , Herpesvirus Humano 1/genética , Humanos
19.
PLoS One ; 6(2): e17267, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21390305

RESUMO

The production of cells capable of expressing gene(s) of interest is important for a variety of applications in biomedicine and biotechnology, including gene therapy and animal transgenesis. The ability to insert transgenes at a precise location in the genome, using site-specific recombinases such as Cre, FLP, and ΦC31, has major benefits for the efficiency of transgenesis. Recent work on integrases from ΦC31, R4, TP901-1 and Bxb1 phages demonstrated that these recombinases catalyze site-specific recombination in mammalian cells. In the present study, we examined the activities of integrases on site-specific recombination and gene expression in mammalian cells. We designed a human artificial chromosome (HAC) vector containing five recombination sites (ΦC31 attP, R4 attP, TP901-1 attP, Bxb1 attP and FRT; multi-integrase HAC vector) and de novo mammalian codon-optimized integrases. The multi-integrase HAC vector has several functions, including gene integration in a precise locus and avoiding genomic position effects; therefore, it was used as a platform to investigate integrase activities. Integrases carried out site-specific recombination at frequencies ranging from 39.3-96.8%. Additionally, we observed homogenous gene expression in 77.3-87.5% of colonies obtained using the multi-integrase HAC vector. This vector is also transferable to another cell line, and is capable of accepting genes of interest in this environment. These data suggest that integrases have high DNA recombination efficiencies in mammalian cells. The multi-integrase HAC vector enables us to produce transgene-expressing cells efficiently and create platform cell lines for gene expression.


Assuntos
Células/metabolismo , Cromossomos Artificiais Humanos/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Integrases/metabolismo , Mutagênese Insercional/métodos , Animais , Células CHO , Células Cultivadas , Cromossomos Artificiais Humanos/metabolismo , Clonagem Molecular/métodos , Cricetinae , Cricetulus , Vetores Genéticos/metabolismo , Humanos , Integrases/genética , Integrases/fisiologia , Modelos Biológicos , Mutagênese Insercional/genética , Organismos Geneticamente Modificados , Recombinação Genética/fisiologia , Transgenes/genética
20.
Methods Mol Biol ; 738: 57-67, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21431719

RESUMO

Intact chromosomes as well as chromosome fragments can be vehicled into various recipient cells without perturbing their ability to segregate as free elements; chromosome transfer can be performed both in cultured cells and in living animals. The method of choice to shuttle single chromosomes between cells is microcell fusion named microcell mediated chromosome transfer (MMCT). The use of MMCT is mandatory in a number of applications where alternative chromosome transfection procedures are ineffective; however, the main drawback is the extremely low efficiency of the technique. Recently, we developed a new procedure to shuttle an engineered human minichromosome from a Chinese hamster ovary hybrid cell line to a mouse embryonic stem cell line. This technology ultimately consists in micronucleated whole cell fusion (MWCF) without microcell isolation. Therefore, MWCF is much more simple than MMCT; moreover, chromosome transfer efficiency is higher. The main limit of the MWCF approach is that it can be employed only with parental cells of different species, while the MMCT protocol can be adapted to any donor and recipient cell line. This chapter will describe both the protocols that we currently use for MMCT and MWCF. The efficiency of the two protocols strictly depends on the parental cell lines to be used for cell fusion.


Assuntos
Fusão Celular/métodos , Cromossomos Artificiais Humanos/metabolismo , Técnicas de Transferência de Genes , Animais , Linhagem Celular , Cricetinae , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Células Híbridas/citologia , Células Híbridas/metabolismo , Camundongos , Polietilenoglicóis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...