RESUMEN
Immunogenomic loci remain poorly understood because of their genetic complexity and size. Here, we report the de novo assembly of a cattle genome and provide a detailed annotation of the immunogenomic loci. The assembled genome contains 143 contigs (N50 ~ 74.0 Mb). In contrast to the current reference genome (ARS-UCD1.2), 156 gaps are closed and 467 scaffolds are located in our assembly. Importantly, the immunogenomic regions, including three immunoglobulin (IG) loci, four T-cell receptor (TR) loci, and the major histocompatibility complex (MHC) locus, are seamlessly assembled and precisely annotated. With the characterization of 258 IG genes and 657 TR genes distributed across seven genomic loci, we present a detailed depiction of immune gene diversity in cattle. Moreover, the MHC gene structures are integrally revealed with properly phased haplotypes. Together, our work describes a more complete cattle genome, and provides a comprehensive view of its complex immune-genome.
Asunto(s)
Genoma , Genómica , Bovinos , Animales , Genoma/genética , Complejo Mayor de Histocompatibilidad/genética , Inmunoglobulinas , Genes de InmunoglobulinasRESUMEN
BACKGROUND: Gene knockout and knock-in have been widely performed in large farm animals based on genome editing systems. However, many types of precise gene editing, including targeted deletion, gene tagging, and large gene fragment replacement, remain a challenge in large farm animals. RESULTS: Here, we established versatile self-excising gene-targeting technology in combination with programmable nucleases (SEGCPN) to efficiently generate various types of precise gene editing in bovine. First, we used this versatile method to successfully generate bovine embryos with point mutations and 11-bp deletions at the MSTN locus. Second, we successfully generated bulls with EGFP labeling at the SRY locus. Finally, we successfully generated humanized cows in which the endogenous 18-kb α-casein gene was replaced with a 2.6-kb human α-lactalbumin gene. CONCLUSIONS: In summary, our new SEGCPN method offers unlimited possibilities for various types of precise gene editing in large animals for application both in agriculture and disease models.
Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Femenino , Animales , Bovinos/genética , Masculino , Humanos , Edición Génica/métodos , Marcación de Gen/métodos , Técnicas de Inactivación de Genes , Mutación PuntualRESUMEN
BACKGROUND: Insulin-like growth factor 2 is a growth-promoting factor that plays an important role in the growth and development of mammals. A nucleotide substitution in intron 3 of IGF2-which disrupts the ZBED6-binding site-affects muscle mass, organ size, and fat deposition in pigs. The ZBED6-binding site is also conserved in cattle. METHODS: In the present study, we introduced mutations in the ZBED6-binding site in intron3 of IGF2 in bovine fetal fibroblasts using the CRISPR/Cas9 system, and investigated the effect of disruption of ZBED6 binding on IGF2 expression. RESULTS: Eleven biallelic-mutant single-cell clones were established, three of which contained no foreign DNA residues. Single-cell clones 93 and 135 were used to produce cloned embryos. Dual-luciferase reporter assay in C2C12 cells demonstrated that the mutation in the ZBED6-binding site increases the promoter 3 activity of bovine IGF2. A total of 49 mutant cloned embryos were transplanted into surrogate cows. Unfortunately, all cloned embryos died before birth. IGF2 was found to be hypomethylated in the only fetus born (stillborn), which may have been due to the incomplete reprogramming. CONCLUSIONS: We efficiently constructed IGF2-edited cell lines and cloned embryos, which provided a theoretical basis and experimental materials for beef cattle breeding.
Asunto(s)
Sistemas CRISPR-Cas , Mamíferos , Animales , Sitios de Unión , Bovinos , Femenino , Intrones/genética , Mamíferos/genética , Mutación , Regiones Promotoras Genéticas , PorcinosRESUMEN
Functional elucidation of bovine Y-chromosome genes requires available genome editing technologies. Meanwhile, it has yet to be proven whether the bovine Sry gene is the main or single factor involved in the development of the male phenotype in bovine. Here, we efficiently knocked out four Y-linked genes (Sry, ZFY, DDX3Y, and EIF2S3Y) in bovine fetal fibroblasts (BFFs) with transcription activator-like effector nucleases (TALENs) individually. Furthermore, we used TALEN-mediated gene knockin at the Sry gene and generated a sex-reversal bovine by somatic cell nuclear transfer (SCNT). The resulting bovine had only one ovary and was sterile. We demonstrate, for the first time, that the Sry gene is an important sex-determining gene in bovine. Our method lays a solid foundation for detecting the biology of the bovine Y chromosome, as it may provide an alternative biological model system for the study of mammalian sex determination, and new methods for the practical application in agricultural, especially for sex predetermination.
Asunto(s)
Técnicas de Sustitución del Gen/métodos , Técnicas de Transferencia Nuclear , Diferenciación Sexual , Proteína de la Región Y Determinante del Sexo/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Cromosoma Y/genética , Animales , Secuencia de Bases , Bovinos , Femenino , Masculino , Homología de Secuencia , Procesos de Determinación del Sexo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genéticaRESUMEN
The function of prolactin in mammary gland development and lactation has been demonstrated in previous studies. However, the potential action of prolactin on mammary duct morphogenesis at mammary anlagen stage (E13.5Ë15.5) has not been elucidated. Transplantation of mammary cells or tissue is an effective approach for the regeneration of damaged mammary gland. Defining the actions of prolactin stimulation on embryonic mammary anlagen has important implications for defining the role of prolactin in the poorly understood processes of mammary duct morphogenesis and regeneration. Here, we have employed the GFP-transgenic mouse model to examine the effects of prolactin on stem cell activity. Using in vitro prolactin stimulation and transplantation of E14.5 mouse mammary anlagen, we identified the influence of prolactin on their mammary repopulating ability. Furthermore, using immunofluorescence staining and microarray analysis, the influence of prolactin on cell characteristics and gene expression in mammary anlagen were also profiled. We also demonstrate that E14.5 mouse mammary anlagen possess mammary stem cells (MaSCs) which achieve mammary repopulation. Furthermore, we found that in vitro prolactin stimulation elevates MaSC activity and promotes the mammary repopulating ability of mammary anlagen. Finally, microarray and pathway analysis showed that MAPK, Akt and JAK-STAT signaling was activated by prolactin stimulation, and drove the mammary repopulation of mammary anlagen. We conclude that prolactin stimulation elevates the stem cell-dependent mammary repopulating ability of embryonic mammary anlagen.
Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/citología , Glándulas Mamarias Animales/citología , Morfogénesis , Células Madre Embrionarias de Ratones/citología , Prolactina/farmacología , Animales , Embrión de Mamíferos/efectos de los fármacos , Femenino , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ratones SCID , Células Madre Embrionarias de Ratones/efectos de los fármacosRESUMEN
The whey protein ß-lactoglobulin (BLG) is a major milk allergen which is absent in human milk. Here, we for the first time generated DNA-free BLG bi-allelic knockout cow by zinc-finger nuclease (ZFNs) mRNA and produced BLG-free milk. According to the allergenicity evaluation of BLG-free milk, we found it can trigger lower allergic reaction of Balb/c mice including the rectal temperature drop and the allergen-specific immunoglobulin IgE production; BLG free-milk was easily digested by pepsin at 2 min, while BLG in control milk was still not completely digested after 60 min, and the binding of IgE from cow's milk allergy (CMA) patients to BLG free-milk was significantly lower than that to the control milk. Meanwhile, the genome sequencing revealed that our animal is free of off-target events. Importantly, editing animal genomes without introducing foreign DNA into cells may alleviate regulatory concerns related to foods produced by genome edited animals. Finally, the ZFNs-mediated targeting in cow could be transmitted through the germline by breeding. These findings will open up unlimited possibilities of modifying milk composition to make it more suitable for human health and also improve the functional properties of milk.
Asunto(s)
Alérgenos/inmunología , Lactoglobulinas/genética , Hipersensibilidad a la Leche/prevención & control , Leche/metabolismo , ARN Mensajero/genética , Nucleasas con Dedos de Zinc/genética , Animales , Bovinos , Femenino , Técnicas de Inactivación de Genes , Humanos , Inmunoglobulina E/metabolismo , Lactoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Leche/química , Hipersensibilidad a la Leche/inmunología , Hipersensibilidad a la Leche/metabolismo , MutaciónRESUMEN
The monoclonal antibody (mAb) against CD20 known as Rituxan is widely used to treat autoimmune diseases and lymphomas. However, further application of Rituxan faces challenges of high production cost, which limits its availability in developing countries. Here, we report a new approach for large production of a recombinant anti-CD20 mAb in the milk of transgenic cattle (at a yield of up to ~6.8 mg/mL), with ~80% recovery rate and >99% purity. Crystallography study showed that our recombinant mAb is structurally nearly identical to Rituxan with only minor differences in N-linked glycosylation pattern. Functional study showed that, while our mAb shared similar target-cell binding capacities and complement-dependent cytotoxicity with Rituxan, our product exhibited a higher binding affinity for FcγRIIIα and a greater antibody-dependent cellular cytotoxicity. Accordingly, our recombinant mAb demonstrated a superior efficacy over Rituxan against B-cell lymphomas in severe combined immunodeficiency mice. Taken together, our data supports transgenic cattle as a novel model for cost-competitive, large-scale production of therapeutic antibodies.
Asunto(s)
Animales Modificados Genéticamente/genética , Anticuerpos Monoclonales/genética , Biotecnología/métodos , Bovinos/genética , Rituximab/genética , Animales , Animales Modificados Genéticamente/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/inmunología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Bovinos/inmunología , Femenino , Expresión Génica , Glicosilación , Linfoma de Células B/tratamiento farmacológico , Ratones SCID , Leche/inmunología , Leche/metabolismo , Rituximab/química , Rituximab/inmunología , Rituximab/uso terapéuticoRESUMEN
To explore the anti-inflammatory activity of endogenous produced melatonin, a melatonin-enriched animal model (goat) with AANAT transfer was successfully generated with somatic cell nuclear transfer (SCNT) technology. Basically, a pIRES2-EGFP-AANAT expression vector was constructed and was transferred into the female fetal fibroblast cells (FFCs) via electrotransfection and then the nuclear of the transgenic FFC was transferred to the eggs of the donor goats. The peripheral blood mononuclear cells (PBMCs) of the transgenic offspring expressed significantly higher levels of AANAT and melatonin synthetic function than those PBMCs from the wild-type (WT) animals. After challenge with lipopolysaccharide (LPS), the transgenic PBMCs had increased autophagosomes and LC3B expression while they exhibited suppressed production of the proinflammatory cytokines, IL1B and IL12 (IL12A-IL12B/p70), compared to their WT. The mechanistic analysis indicated that the anti-inflammatory activity of endogenous melatonin was mediated by MTNR1B (melatonin receptor 1B). MTNR1B stimulation activated the MAPK14 signaling pathway to promote cellular macroautophagy/autophagy, thus, suppressing the excessive inflammatory response of cellular. However, when the intact animals challenged with LPS, the serum proinflammatory cytokines were significantly higher in the transgenic goats than that in the WT. The results indicated that endogenous melatonin inhibited the MAPK1/3 signaling pathway and ROS production, subsequently downregulated gene expression of BECN1, ATG5 in PMBCs and then suppressed the autophagy activity of PBMCs and finally elevated levels of serum proinflammatory cytokines in transgenic animals, Herein we provided a novel melatonin-enriched animal model to study the potential effects of endogenously produced melatonin on inflammatory responses and autophagy activity.
Asunto(s)
N-Acetiltransferasa de Arilalquilamina/genética , Autofagia/genética , Inflamación/genética , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Células Cultivadas , Citocinas/metabolismo , Femenino , Cabras , Inflamación/metabolismo , Leucocitos Mononucleares/metabolismo , Melatonina/metabolismo , Embarazo , Transducción de Señal/genética , Regulación hacia Arriba/genéticaRESUMEN
The genetic modification of cattle has many agricultural and biomedical applications. However, random integration often results in the unstable expression of transgenes and unpredictable phenotypes. Targeting genes to the "safe locus" and stably expressing foreign genes at a high level are desirable methods for overcoming these hurdles. The Rosa26 locus has been widely used to produce genetically modified animals in some species expressing transgenes at high and consistent levels. For the first time, we identified a bovine orthologue of the mouse Rosa26 locus through a genomic sequence homology analysis. According to 5' rapid-amplification of cDNA ends (5'RACE), 3' rapid-amplification of cDNA ends (3'RACE), reverse transcription PCR (RT-PCR) and quantitative PCR (Q-PCR) experiments, this locus encodes a long noncoding RNA (lncRNA) comprising two exons that is expressed ubiquitously and stably in different tissues. The bovine Rosa26 (bRosa26) locus appears to be highly amenable to transcription activator-like effector nucleases (TALENs)-mediated knock-in, and ubiquitous expression of enhanced green fluorescent protein (EGFP) inserted in the bRosa26 locus was observed in various stages, including cells, embryos, fetus and cattle. Finally, we created a valuable master bRosa26-EGFP fetal fibroblast cell line in which any gene of interest can be efficiently introduced and stably expressed using recombinase-mediated cassette exchange (RMCE). The new tools described here will be useful for a variety of studies using cattle.
Asunto(s)
Animales Modificados Genéticamente/genética , Sitios Genéticos/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Animales , Bovinos , Línea Celular , Fibroblastos/metabolismo , ARN no Traducido/genética , Transgenes/genéticaRESUMEN
Stem cell biology offers promise for understanding the origins of the mammary gland. However, the distribution of mammary stem cell (MaSC) activities at earlier embryonic stages has not been fully identified. The markers for sorting adult MaSC, CD24, CD29, and CD49f have been applied to analyze fetal MaSCs. Here we explored mammary anlagen MaSCs by investigating the expression of CD24 and CD49f. According to the comparative analysis between adult mammary gland and fetal mammary anlagen, we found that fetal mouse mammary anlagen may possess a high percentage of potential MaSCs. Flow cytometry analysis revealed 2 distinct mammary anlagen populations: Lin-CD24med and Lin-CD24high. Sphere-forming and mammary repopulating assays confirmed that the stem cell activity of E14.5 mouse mammary anlagen was restricted to the Lin-CD24med cell population. Furthermore, CD24med mammary anlagen cells were separated into Lin-CD24medCD49f+ and Lin-CD24medCD49f- populations and identified, respectively. The results proved that the mammary anlagen Lin-CD24medCD49f+ cell population possesses more stem cell activities than the Lin-CD24medCD49f- cell population. However, a limited numbers of stem cells and large numbers of stromal cells were identified in mammary anlagen in the Lin-CD24med cell population.
Asunto(s)
Antígeno CD24/metabolismo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina alfa6/metabolismo , Glándulas Mamarias Animales/embriología , Células Madre Embrionarias de Ratones/metabolismo , Animales , Embrión de Mamíferos/citología , Femenino , Glándulas Mamarias Animales/citología , Ratones , Células Madre Embrionarias de Ratones/citologíaRESUMEN
Human lactoferrin (hLF) is a valuable protein for pharmaceutical products and functional foods, and worldwide demand for this protein has steadily increased. However, large-scale recombinant human lactoferrin (rhLF) production using current animal bioreactor techniques is limited by the low expression of foreign proteins, the use of antibiotic resistance genes and the down-regulation of endogenous milk proteins. Here, we generated a herd of marker-free, hLF bacterial artificial chromosome (BAC) transgenic cloned cows, as confirmed by Polymerase chain reaction, Southern blot and Western blot analyses. These transgenic cloned cows produced rhLF in milk at concentrations of 4.5-13.6 g/L. Moreover, the total protein content of the milk was increased. Over two hundred transgenic cloned cows were propagated by multiple ovulation and embryo transfer (MOET). A total of 400-450 g of rhLF protein, which shows similar enzymatic activity to natural hLF in iron binding and release, can be purified on a large scale from >100 L of milk per day. Our results suggested that transgenic bovine mammary bioreactors have the potential for large-scale protein production.
Asunto(s)
Expresión Génica , Lactoferrina/biosíntesis , Lactoferrina/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Animales , Animales Modificados Genéticamente , Reactores Biológicos , Bovinos , Orden Génico , Vectores Genéticos , Humanos , Hierro/metabolismo , Lactoferrina/aislamiento & purificación , Leche/metabolismo , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Unión Proteica , Proteínas Recombinantes/aislamiento & purificación , TransfecciónRESUMEN
Bile salt-stimulated lipase (BSSL) is a lipolytic digestive enzyme with broad substrate specificity secreted from exocrine pancreas into the intestinal lumen in all species and from the lactating mammary gland into the milk of some species, notably humans but not cows. BSSL in breast milk facilitates digestion and absorption of milk fat and promotes growth of small for gestational age preterm infants. Thus, purified recombinant human BSSL (rhBSSL) can be used for treatment of patients with fat malabsorption and expressing rhBSSL in the milk of transgenic cloned cows would therefore be a mean to meet a medical need. In the present study, a vector pBAC-hLF-hBSSL was constructed, which efficiently expressed active rhBSSL in milk of transgenic cloned cows to a concentration of 9.8 mg/ml. The rhBSSL purified from cow milk had the same enzymatic activity, N-terminal amino acid sequence, amino acid composition and isoelectric point and similar physicochemical characteristics as human native BSSL. Our study supports the use of transgenic cattle for the cost-competitive, large-scale production of therapeutic rhBSSL.
Asunto(s)
Lipasa/aislamiento & purificación , Animales , Animales Modificados Genéticamente , Western Blotting , Bovinos , Clonación de Organismos , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Calor , Humanos , Concentración de Iones de Hidrógeno , Cinética , Lipasa/química , Lipasa/genética , Lipasa/uso terapéutico , Síndromes de Malabsorción/tratamiento farmacológico , Reacción en Cadena de la PolimerasaRESUMEN
Human lysozyme is an important natural non-specific immune protein that is highly expressed in breast milk and participates in the immune response of infants against bacterial and viral infections. Considering the medicinal value and market demand for human lysozyme, an animal model for large-scale production of recombinant human lysozyme (rhLZ) is needed. In this study, we generated transgenic cloned cows with the marker-free vector pBAC-hLF-hLZ, which was shown to efficiently express rhLZ in cow milk. Seven transgenic cloned cows, identified by polymerase chain reaction, Southern blot, and western blot analyses, produced rhLZ in milk at concentrations of up to 3149.19 ± 24.80 mg/L. The purified rhLZ had a similar molecular weight and enzymatic activity as wild-type human lysozyme possessed the same C-terminal and N-terminal amino acid sequences. The preliminary results from the milk yield and milk compositions from a naturally lactating transgenic cloned cow 0906 were also tested. These results provide a solid foundation for the large-scale production of rhLZ in the future.
Asunto(s)
Animales Modificados Genéticamente/genética , Leche/enzimología , Muramidasa/biosíntesis , Proteínas Recombinantes/biosíntesis , Animales , Animales Modificados Genéticamente/metabolismo , Bovinos , Clonación de Organismos , Fermentación , Humanos , Leche/química , Muramidasa/genética , Proteínas Recombinantes/genéticaRESUMEN
The cells with mammary repopulating capability can achieve mammary gland morphogenesis in a suitable cellular microenvironment. Using cell surface markers of CD24, CD29 and CD49f, mouse mammary repopulating unit (MRU) has been identified in adult mammary epithelium and late embryonic mammary bud epithelium. However, embryonic MRU remains to be fully characterized at earlier mammary anlagen stage. Here we isolated discrete populations of E14.5 mouse mammary anlagen cells. Only Lin(-)CD24(med)CD29(+) cell population was predicted as E14.5 MRU by examining their capacities of forming mammosphere and repopulating cleared mammary fat pad in vivo. However, when we characterized gene expressions of this E14.5 cell population by comparing with adult mouse MRU (Lin(-)CD24(+)CD29(hi)), the gene profiling of these two cell populations exhibited great differences. Real-time PCR and immunostaining assays uncovered that E14.5 Lin(-)CD24(med)CD29(+) cell population was a heterogeneous stroma-enriched cell population. Then, limiting dilutions and single-cell assays also confirmed that E14.5 Lin(-)CD24(med)CD29(+) cell population possessed low proportion of stem cells. In summary, heterogeneous Lin(-)CD24(med)CD29(+) cell population exhibited mammary repopulating ability in E14.5 mammary anlagen, implying that only suitable mammary stroma could enable mammary gland morphogenesis, which relied on the interaction between rare stem cells and microenvironment.
Asunto(s)
Glándulas Mamarias Animales/fisiología , Morfogénesis/genética , Células Madre/fisiología , Animales , Antígeno CD24/genética , Células Epiteliales/fisiología , Femenino , Expresión Génica/genética , Integrina beta1/genética , Masculino , Ratones , Ratones SCIDRESUMEN
Genetically engineered zinc-finger nucleases (ZFNs) are useful for marker-free gene targeting using a one-step approach. We used ZFNs to efficiently disrupt bovine myostatin (MSTN), which was identified previously as the gene responsible for double muscling in cattle. The mutation efficiency of bovine somatic cells was approximately 20%, and the biallelic mutation efficiency was 8.3%. To evaluate the function of the mutated MSTN locus before somatic cell nuclear transfer, MSTN mRNA and protein expression was examined in four mutant cell colonies. We generated marker-gene-free cloned cattle, in which the MSTN biallelic mutations consisted of a 6-bp deletion in one of the alleles and a 117-bp deletion and 9-bp insertion in the other allele, resulting in at least four distinct mRNA splice variants. In the MSTN mutant cattle, the total amount of MSTN protein with the C-terminal domain was reduced by approximately 50%, and hypertrophied muscle fibers of the quadriceps and the double-muscled phenotype appeared at one month of age. Our proof-of-concept study is the first to produce MSTN mutations in cattle, and may allow the development of genetically modified strains of double-muscled cattle.
Asunto(s)
Secuencia de Bases , Desoxirribonucleasas/química , Fibroblastos , Miostatina/genética , Ingeniería de Proteínas , Eliminación de Secuencia , Dedos de Zinc , Animales , Bovinos , Células Cultivadas , Desoxirribonucleasas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genéticaRESUMEN
The PRNP gene encodes a cellular protein named prion, whose misfolded form has been implicated in a number of neuropathic diseases in mammals such as the Bovine Spongiform Encephalopathy (BSE) in cattle. BSE has brought devastating impact on the world economy and human health. Recently, several groups have performed the gene targeting strategy to disrupt the PRNP gene in bovine fibroblast cells and produce BSE-resistant cattle by somatic cell nuclear transfer (SCNT). However, the enrichment efficiency of the gene targeting vector was low. Here, we constructed a novel promoterless gene targeting vector to sequentially disrupt the PRNP gene in bovine fibroblast cells and generate gene targeted cattle by SCNT. The enrichment efficiency of the novel vector was 100% and 60%, respectively. After nuclear transfer, no significant difference was found in the rate of cleavage and blastocyst formation between the knockout and wild type cloned embryos. One PRNPâº/â» calf was born with no obvious abnormal development by now. Fusion RT-PCR and real-time PCR showed one allele of the PRNP gene was functionally disrupted, and the mRNA expression reduced dramatically in the PRNPâº/â» cattle. The reconstituted PRNPâ»/â» embryos showed double alleles disruption, and no difference in the rate of cleavage and blastocyst formation.
Asunto(s)
Bovinos/genética , Clonación de Organismos/métodos , Encefalopatía Espongiforme Bovina/genética , Marcación de Gen , Vectores Genéticos , Priones/genética , Alelos , Animales , Células Cultivadas , Fibroblastos/metabolismo , Genotipo , Humanos , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , TransfecciónRESUMEN
BACKGROUND: Although it is known that RNA interference (RNAi) targeting viral genes protects experimental animals, such as mice, from the challenge of Foot-and-mouth disease virus (FMDV), it has not been previously investigated whether shRNAs targeting FMDV in transgenic dairy cattle or primary transgenic bovine epithelium cells will confer resistance against FMDV challenge. PRINCIPAL FINDING: Here we constructed three recombinant lentiviral vectors containing shRNA against VP2 (RNAi-VP2), VP3 (RNAi-VP3), or VP4 (RNAi-VP4) of FMDV, and found that all of them strongly suppressed the transient expression of a FLAG-tagged viral gene fusion protein in 293T cells. In BHK-21 cells, RNAi-VP4 was found to be more potent in inhibition of viral replication than the others with over 98% inhibition of viral replication. Therefore, recombinant lentiviral vector RNAi-VP4 was transfected into bovine fetal fibroblast cells to generate transgenic nuclear donor cells. With subsequent somatic cell cloning, we generated forty transgenic blastocysts, and then transferred them to 20 synchronized recipient cows. Three transgenic bovine fetuses were obtained after pregnant period of 4 months, and integration into chromosome in cloned fetuses was confirmed by Southern hybridization. The primary tongue epithelium cells of transgenic fetuses were isolated and inoculated with 100 TCID(50) of FMDV, and it was observed that shRNA significantly suppressed viral RNA synthesis and inhibited over 91% of viral replication after inoculation of FMDV for 48 h. CONCLUSION: RNAi-VP4 targeting viral VP4 gene appears to prevent primary epithelium cells of transgenic bovine fetus from FMDV infection, and it could be a candidate shRNA used for cultivation of transgenic cattle against FMDV.
Asunto(s)
Células Epiteliales/citología , Virus de la Fiebre Aftosa/genética , Fiebre Aftosa/virología , ARN Interferente Pequeño/genética , Animales , Animales Modificados Genéticamente , Bovinos , Cricetinae , Fiebre Aftosa/prevención & control , Células HEK293 , Humanos , Lentivirus/genética , Masculino , Oocitos/metabolismo , Interferencia de ARN , Lengua/metabolismo , Replicación ViralAsunto(s)
Endonucleasas/metabolismo , Lactoglobulinas/genética , Animales , Bovinos , Reparación del ADN por Unión de Extremidades , Análisis Mutacional de ADN , Endonucleasas/genética , Eliminación de Gen , Técnicas de Inactivación de Genes , Lactoglobulinas/metabolismo , Mutagénesis Insercional , Fenotipo , Dedos de ZincRESUMEN
Mutations in the PKD2 gene cause autosomal dominant polycystic kidney disease (ADPKD), a common, inherited disease that frequently leads to end-stage renal disease (ESRD). Swine show substantial similarity to humans physiologically and anatomically, and are therefore a good model system in which to decipher the structure and function of the PKD2 gene and to identify potential therapeutic targets. Here we report the cloning and characterization of the porcine PKD2 cDNA showing that the full-length gene (3370 bases) is highly expressed in kidney, with minimal expression in the liver. RNA interference (RNAi) is a promising tool to enable identification of the essential components necessary for exploitation of the pathway involved in cellular processes. We therefore designed four shRNAs and nine siRNAs targeting the region of the porcine PKD2 gene from exons 3 to 9, which is supposed to be a critical region contributing to the severity of ADPKD. The results from HeLa cells with the dual-luciferase reporter system and porcine kidney cells (LLC-PK1) showed that sh12 could efficiently knock down the PKD2 gene with an efficiency of 51% and P1 and P2 were the most effective siRNAs inhibiting 85% and 77% respectively of PKD2 expression compared with untreated controls. A subsequent functional study of the transient receptor potential polycystic (TRPP) 2 channel protein indicated that the decreased expression of TRPP2 induced by siRNA P1 and P2 could release the arrest of the cell cycle from G0/G1 promoting progression to S and G2 phases. Our data, therefore, provides evidence of potential knock-down target sites in the PKD2 gene and paves the way for the future generation of transgenic ADPKD knock-down animal models.