Open Access

The goat β‑casein/CMV chimeric promoter drives the expression of hLF in transgenic goats produced by cell transgene microinjection

  • Authors:
    • Ting Zhang
    • Yuguo Yuan
    • Rui Lu
    • Sheng Xu
    • Minya Zhou
    • Tingting Yuan
    • Yaoyao Lu
    • Kunning Yan
    • Yong Cheng
  • View Affiliations

  • Published online on: October 24, 2019     https://doi.org/10.3892/ijmm.2019.4382
  • Pages: 2057-2064
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

There is growing interest in the application of lactoferrin (LF) as a drug or food additive for animals and humans. The objective of this study was to produce transgenic cloned goats that would serve as living bioreactors, expressing high levels of recombinant human LF (rhLF) in their milk. We designed a pCL25 expression vector containing goat β‑casein/CMV chimeric promoter in order to facilitate rhLF expression. This pCL25‑rhLF‑Neo vector was microinjected into goat fetal fibroblasts. G418 selection and PCR analysis were used to identify transgenic donor cells suitable for somatic cell nuclear transfer (SCNT). After SCNT and embryo transplantation, goats harboring the hLF gene were produced, as confirmed via PCR and southern blotting. The average rhLF concentration in milk from this transgenic goat was 3.89 mg/ml as determined via ELISA. We also used an optimized buffer in order to effectively elute high‑purity (95.8%) rhLF from a cation‑exchange column, with the recovered rhLF exhibiting high biological activity. Findings from this study demonstrated that it is possible to generate a transgenic goat harboring the hLF transgene driven by the goat β‑casein/CMV chimeric promoter. It represents an initial step towards the production of rhLF, potentially allowing for industrialized purification in the future.

Introduction

There is an increasing need for the production of recombinant therapeutic proteins generated via a range of transgenic techniques, with the optimal approach to such expression being to produce these recombinant proteins in bioreactors such as bacteria, yeast, plants, mammalian cells, or transgenic animals (1-7). Among these bioreactors, mammary gland bioreactors in transgenic animals offer the advantage of being fully compatible with humans and being approved by the FDA (8). Such mammary gland-derived recombinant proteins have already been implemented for clinical use (9) For example, recombinant human antithrombin III (Atryn®) is produced from the milk of transgenic goats (10). Such mammary gland bioreactors are highly advantageous for the production of those proteins that require post-translational modifications in order to mediate their stability or activity (11). Producing recombinant proteins in mammary glands therefore represents a more profitable approach to the production of recombinant human proteins. Pantano et al suggested that relatively few mammary cells in transgenic animals ultimately express recombinant proteins (12), underscoring that there is an urgent need to determine how to bolster the in vivo expression of these recombinant proteins using optimized expression vector systems.

Achieving high-level production of recombinant proteins within the milk of transgenic animals depends upon ensuring high-level transcription of the introduced cDNA. This makes it essential to select appropriate cis-acting elements, including promoters and enhancers, for the introduced genes. Large quantities of β-casein proteins are produced in goats during lactation in response to hormonal stimulation with β-casein concentrations being 43% higher in goat milk relative to bovine milk (13,14). It is thought to be a binding site for STAT5a in the -300 bp region of the goat β-casein promoter, and this binding site serves to mediate responses to lactogenic hormone stimulation (15). This lays the theoretical foundation for the selection of a goat β-casein promoter, thus allowing for the efficient expression of proteins in mammary glands.

Although the goat β-casein promoter has previously been widely used to drive the transcription of many recombinant proteins in transgenic goats, the expression of these proteins has not been sufficiently high for commercial applications. A variety of approaches have been employed in an effort to boost mammary expression of these recombinant proteins, including the use of distal regulatory elements/large genomic DNA fragments (16), insulators (17), matrix-attached regions (18), and targeted site integrations (19). The cytomegalovirus (CMV) promoter is a high-efficiency promoter/enhancer widely used for transgene expression in cells. Zarrin et al found this promoter to be more efficient than alternatives such as the SV40, Rous sarcoma virus (RSV), and Vλl promoters for certain B-cell lines (20). There are few reports, however, regarding the use of a goat β-casein/CMV chimeric promoter to facilitate protein production in the mammary glands of medium and large transgenic animals.

The properties of a given protein determine its protein purification strategy. LF is a cationic protein, thus making it well suited to purification via cation-exchange chromatography (21,22). This approach is widely used for bovine LF purification by bLF-producing companies. Concanavalin A affinity chromatography or metal ion affinity chromatography are also viable strategies for purifying LF owing to its glycosylation and Fe3+-binding activity (23,24).

In this study, we generated a transgenic goat harboring the human lactoferrin transgene driven by a chimeric goat β-casein/CMV promoter. This animal was generated using goat fetal fibroblasts microinjected with the pCL25-rhLF-neo vector as SCNT donor cells, allowing for mammary gland-specific transgene expression (25,26), while retaining the biological characteristics necessary for better efficacy as a drug or food additive. We additionally conducted ELISAs, western blot, and antibacterial activity assays to confirm that human lactoferrin was efficiently expressed in transgenic goat milk while retaining its normal biological activity.

Materials and methods

Ethics statement

Animal experiments and procedures were performed in accordance with the guide for the Care and Use of Laboratory Animals (Ministry of Science and Technology of the People's Republic of China) and approved by the animal care and use committee of Yangzhou University, Yangzhou, China [license no. SYXK(Su)2017-0044]. A total of 50 female dairy goats (45-60 kg, 13-18 months old; Jiangsu Academy of Agricultural Sciences, Nanjing, China) used in the current study were raised at room temperature (25±2°C), with a 12 h day/night cycle, and allowed free access to food and water. All animals were anesthetized using xylazine hydrochloride injection (0.001-0.002 ml/kg) purchased from huamu Animal health Products Co., Ltd. during surgery, with all possible effort being made to reduce their pain, distress, and suffering.

Lactoferrin expression vector construction

Human lactoferrin (GenBank: KT006756.1) cDNA was synthesized by Genscript (China), using cDNA containing 5′ and 3′ terminal XhoI sites. The sequence encoding the mature lactoferrin peptide was fused to both the goat β-lactoglobulin signal peptide as well as the Kozak translation initiation sequence.

The synthesized lactoferrin gene was cloned into the pCL25 vector (generated internally) containing the goat β-casein/CMV chimeric promoter and a neo-selectable cassette in the goat β-casein 3′ genomic region near the vector NotI site (Fig. 1). NotI and SalI were used for vector digestion, and a QIAquick Gel Extraction kit (28704, Qiagen, Germany) was used to purify the resultant fragments.

Cell culture and transgene expression

A 30-day old fetus was surgically removed from a Sannen dairy goat and used to generate fibroblasts. Briefly, fetal tissue was cut into small fragments following the removal of the internal organs, head, and limbs, and these fragments underwent 0.05% trypsin-EDTA-mediated digestion. Fibroblasts were then isolated from the supernatant portion of this digestion and grown using DMEM/F12 (Sh30023.01; hyclone) containing 10% FBS (Sh30406.01; hyclone), and 1% penicillin-streptomycin (SV30010; hyclone) at 37°C in a 5% Co2 humidified incubator. Cells underwent passaging at 80% confluency, and after the second passage cells were aliquoted and frozen in freezing media containing 10% DMSo (D2650; Sigma) and 20% FBS.

Aliquots of cells were frozen, and once they grew to 80% confluency they were microinjected with 5 ng/µl of the purified pCL25-rhLF-Neo DNA fragment using an Eppendorf InjectMan (NI2; Eppendorf) and then cultured as above. After 24 h, the cells were grown in selective medium containing 800 ng/µl G418 (SV3006801; hyclone) for approximately 10 days. A cloning ring was used to isolate and expand healthy colonies following selection, and these clones were subcultured as above. Some of these subcultures were frozen for long-term storage, while the rest were screened for expression of the transgene via polymerase chain reaction (PCR).

Generation of a transgenic goat via SCNT

Somatic cell nuclear transfer (SCNT) was conducted after identifying transgene-positive clones. Enucleated oocytes served as recipients for transgenic cell nuclei, with a super electro cell fusion generator (EGFE21; nepa Gene) being used for the SCnT procedure. next, 5 µmol/l ionomycin (I0634; Sigma) and 7.5 µg/ml cytochalasin B (C6762; Sigma) in M16 medium (M7292; Sigma) was used to activate these reconstructed embryos for 5 min, and then cells were treated with M16 containing 2 mmol/l 6-dimethylaminopurine (D2629; Sigma) and 7.5 µg/ml cytochalasin B for 5 h. After activation, the embryos were implanted into recipient goats, and after a 1-month period these animals were assessed via ultrasound to confirm pregnancy. Approximately 150 days later, kids were delivered naturally. For all kids, a small portion of the ear was taken as a biopsy sample from which DNA was isolated and used to assess transgene incorporation by PCR and southern blotting. DL2000 DNA marker (3427A; Takara) was purchased from Takara Biotech (Dalian) Co., Ltd.

Confirmation of transgene integration in cloned goats

Genomic DNA from the transgenic donor cells and ear tissues of cloned goats was prepared with an Easy Pure Genomic DNA kit (EE101-1; TransGen). A pair of primers specific for human LF was used to determine which donor cells had incorporated the transgene. Primer sequences used were: CMV-crhLF-1: ATG GGC GTG GAT AGC GGT TTG AC and CMV-crhLF-2: CCA CCA TCA AGG GTC ACA GCA TCG. To identify transgenic goats, the following primers were instead used: CMV-grhLF-1: ATA GTA ACG CCA ATA GGG A and CMV-grhLF-2: GGT CG CA GTT TGT AGG G. The following conditions were used for all PCR reactions: 94°C for 5 min, 33 cycles at 94°C for 1 min, 56°C for 1 min, then 72°C for 38 sec, and finally held at 72°C for 10 min. Product sizes for the two primer pairs were 450 and 775 bp, respectively. Sequencing analysis was performed by Sangon Biotech (Shanghai) Co., Ltd.

Southern blotting was next employed to confirm specific transgene DNA integration in goats. Ear biopsy-derived DNA from transgenic and wild-type (WT) goats underwent overnight BamHI digestion, with the PCL25-CMV-rhLF-neo plasmid serving as a positive control. A digoxigenin-labeled probe then underwent PCR amplification with the CMV-grhLF-1 and CMV-ghLF-2 primer pair. Samples underwent 4-h agarose gel electrophoresis, after which DNA was transferred to a nylon membrane (11417240001; Roche) for blotting. This membrane next underwent probe hybridization for 18 h, followed by incubation with biotin-labeled mouse anti-Digoxin for 30 min. A positive band was expected to be approximately 9.1 kb in size. Southern blotting reagents were purchased from Boster Co. (Wuhan).

ELISAs

Milk samples collected from lactating transgenic and WT goats were centrifuged at 10,000 × g for 30 min at 4°C for whey isolation. The samples were diluted at 1:10 with PBS, and were used for ELISA reactions with a rabbit-anti-lactoferrin polyclonal primary antibody (dilution 1:2,000, 4% FBS/PBS; Sangon; D121815-0025). After incubation at 37°C for 1 h and being washed three times with PBS-T (PBS containing 0.05% Tween-20), wells were probed with an hRP-conjugated goat-anti-rabbit secondary antibody (dilution 1:1,000, 4% FBS/PBS; Sangon) at 37°C for 1 h. The samples then underwent a colorimetric reaction via adding TMB substrate to each well, after which absorbance at 450 nm was measured via microplate reader (Rayto). Protein standards (SRP6519; Sigma) were used for standard curve generation, and sample rhLF concentrations were determined based on this standard curve.

Purification of rhLF from the transgenic cloned goat

Fat and other undissolved substances were removed from milk via centrifugation at 12,000 × g for 30 min at 4°C, after which the pH was reduced to 4.0 in order to facilitate casein precipitation. Milk was then centrifuged at 4°C at 100,000 × g for 1 h. Supernatant pH was adjusted to 6.0 using acetic acid, after which the samples were centrifuged again as in the previous step. A protein purification system (ÄKTAprimeTM PLuS; GE healthcare) was used for all purification reactions. First, after equilibration in a column containing Buffer A (0.07 mol/l HAc, pH 3.1), samples were loaded onto a HiTrap Capto S cation exchange column (1 ml; GE healthcare) and the bound proteins were eluted via a step gradient of 30 and 100% Buffer B (0.5 mol/l NaCl, 0.07 mol/l Tris-HAc, pH 7.5). The eluate containing 100% Buffer B was collected and desalted via a Bestdex G-25 column (1.6×2.5 cm; BestChrom) for use in downstream experiments. SDS-PAGE analysis was then used to assess protein purity.

Western blotting

Whey was isolated as above and then boiled in SDS loading buffer for 10 min, after which samples were electrophoretically separated using 12% polyacrylamide Tris-glycine gels. Afterwards, these gels were stained using Coomassie Brilliant Blue G-250, and the sample purity and concentrations were determined using Tanon Gis software (Bio-Tanon). For western blotting, separated proteins were then transferred onto PVDF membranes (F019531; Sangon). The membranes were blocked using 5% BSA/TBST overnight at 4°C, and then probed using a polyclonal rabbit-anti-LF antibody (1:2,000, 10% FBS/TBST; Sangon) at 37°C for 1.5 h. next, an hRP-conjugated secondary goat-anti-rabbit IgG (1:1,000, 10% FBS/TBST; Sangon) was used to probe blots at 37°C for 1 h. The blots were then washed three times in TBST (20 mM Tris-base, 137 mM naCl, 0.05% Tween-20), and protein bands were detected with an ECL substrate solution (Millipore Corporation) based on provided directions.

Bacteriostatic activity assessment

Lactoferrin has been shown to be able to inhibit the growth of both Gram-positive and -negative bacteria, including important pathogenic species such as Helicobacter pylori, Staphylococcus aureus, Shigella flexneri, enteropathogenic Escherichia coli (EPEC) and Salmonella enterica serovar typhimurium (2,27-31). We therefore selected E. coli K88 grown on LB plates as a model strain to test the bacteriostatic activity of transgenic goat milk.

A single E. coli K88 colony was transferred into 15 ml LB culture medium, shaken and kept overnight at 37°C. The resultant bacteria were then streaked evenly across an LB agar plate using a cotton swab.

To test bacteriostatic activity of the milk, sterile filter papers (8 mm in diameter) were placed onto the plate surface, and 100 µl sample aliquots were added on top of this paper. After a 4-h incubation at 37°C, the size of the growth inhibition area surrounding a given sample was used to assess bacteriostatic activity.

Results

Lactoferrin expression vector construction

We successfully inserted the LF cDNA fragment into the pCL25 vector, thereby producing a pCL25-rhLF-Neo recombinant vector that was found to be of appropriate size based on restriction enzyme digestion and sequencing. Sequencing confirmed that the rhLF coding region was fused in-frame upstream of pCL25.

Fetal goat fibroblast transfection

Goat fetal fibroblasts were microinjected with 5 ng/µl of the purified pCL25-rhLF-neo DNA fragment, and were selected using G418. A total of 16 G418-resistant transgenic cells were obtained by single cell amplification. of these, 9 were determined to express the hLF transgene via PCR using the CMV-crhLF-1 and CMV-crhLF-2 primers (Fig. 2). In total, 56.25% (9/16) of the cell clones had confirmed pCL25 integration. Clone no. 4 cells served as SCNT donors as they were found to exhibit the best viability and quality.

SCNT-mediated transgenic cloned goat generation

SCNT was used to produce transgenic goats as previously identified (27). We transferred 65 reconstructed embryos into 5 recipient goats, leading to the birth of a single female kid that was found by PCR to harbor the pCL25-rhLF-neo transgene (Table I). The female kid was designated as LF-1 (Fig. 3), and upon reaching sexual maturity underwent mating with a WT ram.

Table I

Nuclear transfer outcomes.

Table I

Nuclear transfer outcomes.

Cell linesOocytesReconstructed embryosTransferred embryosRecipientsPregnancies at day 30No. of newborns
No. 4957065511
Confirmation of transgene integration in cloned goats

PCR and Southern blotting were used to confirm that the transgenic goat integrated the rhLF transgene. human LF-specific primers (CMV-grhLF-1 and CMV-grhLF-2) were used to identify the cloned goats by PCR, while digoxigenin-labeled versions of these primers were used as probes for Southern blotting.

Following PCR, we were able to amplify a 775 bp product, confirming successful rhLF transgene integration into this cloned goat (Fig. 4A). Southern blotting further confirmed this finding (Fig. 4B).

Assessment of milk rhLF expression in transgenic goats

Expression of rhLF in WT and transgenic goat milk samples was next assessed via ELISA. Milk was collected during days 1-30 of lactation following delivery. We found that the rhLF concentration reached a peak of 4.7 mg/ml on day 4, with an average concentration of 3.89±0.82 mg/ml from days 1-30 of lactation.

To assess the possible ectopic expression of rhLF in this transgenic goat, rhLF levels in the serum and saliva of lactating goat were measured via ELISA. There was no indication of rhLF expression in the serum or saliva of this transgenic goat (data not shown).

Purification of rhLF from the transgenic cloned goat

Cation exchange chromatography can be used to separate lactoferrin from milk, as lactoferrin has a net-positive charge. In order to explore the optimal elution conditions for separation and purification of rhLF via cation exchange chromatography, we first assessed the optimal solution conductivity for rhLF. Stepwise elution was used to achieve one-step elution and separation of the target protein.

Two elution peaks were obtained from the HiTrap Capto S cation exchange column eluted with a step gradient of 30 and 100% Buffer B. SDS-PAGE and western blotting revealed that high-purity rhLF was successfully collected in peak P3 of the eluent (Fig. 5), with a size of 80 kDa. The concentration of the purified rhLF was found to be 1.25 mg/ml by spectrophotometry (One Drop 1000+; OneDrop Technologies, Inc.). The purity was determined to be 95.8% based on densitometric scanning of the SDS-PAGE gel.

Western blotting confirmed that samples in the transgenic goat were identical to native hLF control samples, with a size of approximately 80 kDa (Fig. 6). Bands were absent in the WT control goat sample, as expected.

Assessment of transgenic goat milk bacteriostatic activity

The bacteriostatic activity of the rhLF in the transgenic goat milk was assessed via an agar disc diffusion method in order to allow for observation of bacteriostatic activity in vitro. Sterile filter paper was placed onto agar plates containing E. coli K88, and bacteriostatic activity was estimated based on inhibition zone sizes surrounding the sterile filter papers following a 4 h incubation at 37°C. These results revealed that rhLF from transgenic milk exhibited comparable bacteriostatic activity to that of hLF (inhibition zone diameters of 17 and 19 mm, respectively). WT goat milk served as a negative control, with no inhibition zone being evident. We also found that rhLF purified by cation-exchange chromatography exhibited similar bacteriostatic activity (an inhibition zone diameter of 13 mm) (Fig. 7).

Discussion

In this study, we successfully used SCNT as a means of generating a transgenic goat producing rhLF in mammary cells, using transgenic goat fetal fibroblast cells as donor cells. To date, there have been no previous reports of using fetal fibroblasts microinjected with rhLF gene as donor cells for SCNT. We detected no abnormalities in the founder transgenic goat or its offspring, indicating no effect of the vector on goat biology. To determine whether the rhLF transgene could be stably transmitted to offspring, the female founder transgenic goat was mated with a wild-type ram and a single male lamb was birthed. A subsequent PCR assay demonstrated that it was transgenic for rhLF (data not shown), indicating that the rhLF transgene can be inherited by offspring.

There are many reports regarding high-level rhLF expression in transgenic mice, rabbits, and cows (32-34). Mice and rabbits, however, are not suitable for large-scale commercial rhLF production due to their limited milk production and short life span (2-3 years for mice, 8-10 years for rabbits). Cows also are not appropriate models for producing rhLF because bovine milk has more allergenic protein than does goat milk (35,36). Therefore, goats are more suitable as a biologic mammary reactor for the large-scale production of rhLF given that goat milk has been reported to contain smaller fat globules and a distinct casein composition relative to bovine milk, making it less allergenic (37,38). At present, many transgenic animals are produced via SCNT or pronucleus microinjection, including sheep (39), goats (40,41), cows (42), mice (43) and rabbits (44). The success rate of SCNT remains low and varies based upon factors such as the vector used, the source of recipient and donor cells, the exact SCnT protocol employed, and the influence of exogenous genes on embryonic development (45-47). The quality of donor cells is critical for producing transgenic animals via SCnT. Preparation of transgenic animals using electroporation-mediated transfection requires optimization of transfection conditions and is often associated with a high rate of cell death. however, cell microinjection avoids these challenges, instead offering a high integration rate while remaining suitable for genetic engineering and the establishment of transgenic animals. In this study, we improved upon the process of preparing transgenic goats using goat fetal fibroblasts microinjected with the rhLF gene as donor cells for SCNT.

In previous reports, we constructed various mammary gland-specific vectors containing a CMV enhancer and a chimeric promoter [goat β-casein, bovine αs1-casein, and goat β-lactoglobulin (BLG)] based on milk protein promoter sequences. These vectors allowed for 1.17-8.10 mg/ml hLF levels in transgenic murine milk-roughly 100,000-fold higher than the levels produced from control promoters (7-40 ng/ml). We also found that the inclusion of the CMV enhancer significantly increased hLF expression in these mice. Use of hLF cDNA did not achieve expression levels as high as those from hLF genomic DNA in these mice (25). Many factors can influence recombinant milk protein expression levels, including copy number, site of chromosomal insertion, and species-specific differences in expression patterns (48,49). rhLF expression levels in transgenic goats can be as high as 4.7 mg/ml-levels which are markedly higher than the levels observed in transgenic goats without a CMV enhancer (50). There were no indications of rhLF expression in the serum or saliva of the transgenic goat, as the goat β-casein promoter is specifically expressed only in lactating mammary tissue and not at other ectopic sites. This means that there is no potential risk of transgenic animals expressing heterologous proteins in the mammary gland when using this goat β-casein/CMV chimeric promoter. using western blotting we further confirmed that the size of the rhLF expressed in transgenic goats was roughly 80 kDa, which is comparable to the size of hLF.

The secretion of lactoferrin in milk is directly related to the nutritional status and environmental conditions of the mother, and as such the secretion of lactoferrin in milk can be improved by improving maternal housing conditions and other factors. however, for transgenic animals, in addition to these growth conditions and environmental factors, improving the inheritance and stability of foreign genes remains a major challenge. In this study, our aim was to produce transgenic-cloned goats as living mammary bioreactors that exhibited a high-level of rhLF expression in their milk. An optimized construct is essential in order to achieve a high level expression of recombinant proteins. We used ELISA to confirm the expression of rhLF in transgenic goat milk on days 1-30 of lactation following delivery, revealing that rhLF was continuously expressed in goat milk during this 30-day period. There were no clear decreases in rhLF expression during the lactation period. These results thus clearly show that a transgenic goat carrying the pCL25-rhLF-Neo mammary gland-specific expression vector encoding goat β-casein/CMV chimeric promoter can express rhLF stably in the mammary gland.

At present, phosphoric buffer (PB) is widely used as an eluent when extracting lactoferrin via cation-exchange chromatography (23). however, PB easily associates with common Ca2+ ions, Mg2+ ions, and heavy metal ions to form precipitates, and it can also inhibit certain biochemical processes as well as the activity of most enzymes. PB is thus not an ideal eluent choice when purifying lactoferrin by cation-exchange chromatography. In order to achieve superior purified rhLF activity, we therefore used a commercially available HiTrap Capto S cation exchange column for its effective purification from the milk of a transgenic goat, using the Tris-HAc buffer as an eluent. Similarly to the rhLF purified in other previous reports (24,51), rhLF purification efficiency in transgenic goats was high (≥95.8%). When we assessed the bacteriostatic activity of this rhLF, we found it to be comparable to that of natural hLF, penicillin, and streptomycin, which suggests that rhLF may be an effective antibiotic for future use.

In conclusion, we have successfully used SCNT to produce a transgenic goat, with goat fetal fibroblast cells serving as donor cells microinjected with the expression vector pCL25. Our results conclusively demonstrate that the pCL25 vector, which contains goat β-casein/CMV chimeric promoter, can drive transgenic goats to stably express a biologically active form of rhLF. This study offers an initial strategy for rhLF production for incorporation into drugs or food products, thereby facilitating future studies of this protein.

Acknowledgments

We thank all lab members for their help in this study.

Funding

This study was supported by the National Key Research and Development Program of China, China (no. 2016YFE0126000), the Priority Academic Program Development of Jiangsu higher Education Institutions, China (PAPD) and the Yangzhou university-enterprise Cooperation Project, China (YZ2017283).

Availability of data and materials

All data and materials are available from the authors.

Authors' contributions

TZ, YY, RL, SX, MZ, TY, YL, and KY performed the studies and analyzed the data. TZ and YC wrote and revised the manuscript. YC conceived and guided the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The Guide for the Care and use of Laboratory Animals (Ministry of Science and Technology of the People's Republic of China) was followed for all surgical studies described herein, which received prior approval from the Yangzhou University Animal Care and Use Committee (license no. SYXK(Su)2017-0044). All animals were anesthetized during surgery, with all possible effort being made to reduce the pain, distress, and suffering of the study animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

García-Montoya IA, Cendón TS, Arévalo-Gallegos S and Rascón-Cruz Q: Lactoferrin a multiple bioactive protein: An overview. Biochim Biophys Acta. 1820:226–236. 2012. View Article : Google Scholar

2 

Wu J, Hu Y, Du C, Piao J, Yang L and Yang X: The effect of recombinant human lactoferrin from the milk of transgenic cows on Salmonella enterica serovar typhimurium infection in mice. Food Funct. 7:308–314. 2016. View Article : Google Scholar

3 

Fliedl L, Grillari J and Grillari-Voglauer R: human cell lines for the production of recombinant proteins: On the horizon. New Biotechnol. 32:673–679. 2015. View Article : Google Scholar

4 

Fu Z, Baker D, Cheng A, Leighton J, Appelbaum E and Aon J: Characterization of a saccharomyces cerevisiae fermentation process for production of a therapeutic recombinant protein using a multivariate bayesian approach. Biotechnol Prog. 32:799–812. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Gupta SK and Shukla P: Advanced technologies for improved expression of recombinant proteins in bacteria: Perspectives and applications. Crit Rev Biotechnol. 36:1089–1098. 2016. View Article : Google Scholar

6 

Kim H, Yoo SJ and Kang HA: Yeast synthetic biology for the production of recombinant therapeutic proteins. FEMS Yeast Res. 15:1–16. 2015.

7 

Song S, Ge X and Cheng Y, Lu R, Zhang T, Yu B, Ji X, Qi Z, Rong Y, Yuan Y and Cheng Y: high-level expression of a novel recombinant human plasminogen activator (rhPA) in the milk of transgenic rabbits and its thrombolytic bioactivity in vitro. Mol Biol Rep. 43:775–783. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Lavine G: FDA approves first biological product derived from transgenic animal. Am J Health Syst Pharm. 66:5182009. View Article : Google Scholar : PubMed/NCBI

9 

Davis B and Bernstein JA: Conestat alfa for the treatment of angioedema attacks. Ther Clin Risk Manag. 7:265–273. 2011.PubMed/NCBI

10 

The farmyard drug store: Nature. 443:16–17. 2006. View Article : Google Scholar

11 

Bösze Z, Baranyi M and Whitelaw CB: Producing recombinant human milk proteins in the milk of livestock species. Adv Exp Med Biol. 606:357–393. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Pantano T, Rival-Gervier S, Prince S, Menck-Le Bourhis C, Maeder C, Viglietta C, Houdebine LM and Jolivet G: In vitro and in vivo effects of a multimerized alphas 1-casein enhancer on whey acidic protein gene promoter activity. Mol Reprod Dev. 65:262–268. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Huma N, Ghaffar F, Rafiq S, Pasha I, Sameen A, Hayat I and Hussain I: Characterization of milk proteins from different animal species through Gel electrophoresis. Pak J Zool. 50:1983–1986. 2018. View Article : Google Scholar

14 

Dayal R, Hurlimann J, Suard YM and Kraehenbuhl JP: Chemical and immunochemical characterization of caseins and the major whey proteins of rabbit milk. Biochem J. 201:71–79. 1982. View Article : Google Scholar : PubMed/NCBI

15 

Kung MH, Lee YJ, Hsu JT, Huang MC and JU YT: A functional study of proximal goat β-casein promoter and intron 1 in immortalized goat mammary epithelial cells. J Dairy Sci. 98:3859–3875. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Giraldo P and Montoliu L: Size matters: Use of YACs, BACs and PACs in transgenic animals. Transgenic Res. 10:83–103. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Fujiwara Y, Miwa M, Takahashi R, Kodaira K, Hirabayashi M, Suzuki T and Ueda M: high-level expressing YAC vector for transgenic animal bioreactors. Mol Reprod Dev. 52:414–420. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Sharp JA, Cane Kn, Mailer SL, Oosthuizen WH, Arnould JP and Nicholas KR: Species-specific cell-matrix interactions are essential for differentiation of alveoli like structures and milk gene expression in primary mammary cells of the Cape fur seal (Arctocephalus pusillus pusillus). Matrix Biol. 25:430–442. 2006. View Article : Google Scholar : PubMed/NCBI

19 

McKnight RA, Shamay A, Sankaran L, Wall RJ and Hennighausen L: Matrix-attachment regions can impart position- independent regulation of a tissue-specific gene in transgenic mice. Proc Natl Acad Sci USA. 89:6943–6947. 1992. View Article : Google Scholar

20 

Zarrin AA, Malkin L, Fong I, Luk KD, Ghose A and Berinstein NL: Comparison of CMV, RSV, SV40 viral and Vlambda1 cellular promoters in B and T lymphoid and non-lymphoid cell lines. Biochim Biophys Acta. 1446:135–139. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Law BA and Reiter B: The isolation and bacteriostatic properties of lactoferrin from bovine milk whey. J Dairy Res. 44:595–599. 1977. View Article : Google Scholar : PubMed/NCBI

22 

Yoshida S, Wei Z, Shinmura Y and Fukunaga N: Separation of lactoferrin-a and -b from bovine colostrum. J Dairy Sci. 83:2211–2215. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Nuijens JH, van Berkel PH and Schanbacher FL: Structure and biological actions of lactoferrin. J Mammary Gland Biol neoplasia. 1:285–295. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Aguila A, Herrera AG, Toledo W, Rodríguez AA, Garcia HM, Hernández J, Cádiz A, Sierra G and Brock JH: Isolation and structure-functional characterization of human colostral lacto-ferrin. Biotecnol Appl. 17:177–182. 2000.

25 

Cheng Y, An LY, Yuan YG, Wang Y, Du FL, Yu BL, Zhang ZH, Huang YZ and Yang TJ: hybrid expression cassettes consisting of a milk protein promoter and a cytomegalovirus enhancer significantly increase mammary-specific expression of human lactoferrin in transgenic mice. Mol Reprod Dev. 79:573–585. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Yuan YG, An L, Yu B, Song S, Zhou F, Zhang L, Gu Y, Yu M and Cheng Y: Expression of recombinant human alpha-lactalbumin in the milk of transgenic goats using a hybrid pomoter/enhancer. J Anal Methods Chem. 2014:2810312014. View Article : Google Scholar : PubMed/NCBI

27 

Gomez HF, Ochoa TJ, Herrera-Insua I, Carlin LG and Cleary TG: Lactoferrin protects rabbits from Shigella flexneri-induced inflammatory enteritis. Infect Immun. 70:7050–7053. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Ochoa TJ, Brown EL, Guion CE, Chen JZ, McMahon RJ and Cleary TG: Effect of lactoferrin on enteroaggregative E. coli (EAEC). Biochem Cell Biol. 84:369–376. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Ochoa TJ, Noguera-Obenza M, Ebel F, Guzman CA, Gomez HF and Cleary TG: Lactoferrin impairs type III secretory system function in enteropathogenic Escherichia coli. Infect Immun. 71:5149–5155. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Farnaud S and Evans RW: Lactoferrin-a multifunctional protein with antimicrobial properties. Mol Immunol. 40:395–405. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Mosquito S, Ochoa TJ, Cok J and Cleary TG: Effect of bovine lactoferrin in Salmonella ser. Typhimurium infection in mice. Biometals. 23:515–521. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Nuijens JH, van Berkel PH, Geerts ME, Hartevelt PP, de Boer HA, van Veen HA and Pieper FR: Characterization of recombinant human lactoferrin secreted in milk of transgenic mice. J Biol Chem. 272:8802–8807. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Li L, Shen W, Min L, Dong H, Sun Y and Pan Q: human lactoferrin transgenic rabbits produced efficiently using dimethylsulfoxide- sperm-mediated gene transfer. Reprod Fertil Dev. 18:689–695. 2006. View Article : Google Scholar

34 

van Berkel PH, Welling MM, Geerts M, van Veen HA, Ravensbergen B, Salaheddine M, Pauwels EK, Pieper F, Nuijens JH and Nibbering PH: Large scale production of recombinant human lactoferrin in the milk of transgenic cows. Nat Biotechnol. 20:484–487. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Pastuszka R, Barłowska J and Litwińczuk Z: Allergenicity of milk of different animal species in relation to human milk. Postepy Hig Med Dosw (online). 70:1451–1459. 2016. View Article : Google Scholar

36 

Høst A, Husby S, Gjesing B, Larsen JN and Løwenstein H: Prospective estimation of IgG, IgG subclass and IgE antibodies to dietary proteins in infants with cow milk allergy. Levels of antibodies to whole milk protein, BLG and ovalbumin in relation to repeated milk challenge and clinical course of cow milk allergy. Allergy. 47:218–229. 1992. View Article : Google Scholar

37 

Pisanu S, Marogna G, Pagnozzi D, Piccinini M, Leo G, Tanca A, Roggio AM, Roggio T, Uzzau S and Addis MF: Characterization of size and composition of milk fat globules from Sarda and Saanen dairy goats. Small Ruminant Res. 109:141–151. 2013. View Article : Google Scholar

38 

Juvarajah T, Wan-Ibrahim WI, Ashrafzadeh A, Othman S, Hashim OH, Fung SY and Abdul-Rahman PS: Human milk fat globule membrane contains hundreds of abundantly expressed and nutritionally beneficial proteins that are generally lacking in caprine milk. Breastfeed Med. 13:631–637. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Schramm FR: The Dolly case, the Polly drug, and the morality of human cloning. Cad Saude Publica. 15(Suppl 1): 51–64. 1999. View Article : Google Scholar : PubMed/NCBI

40 

An LY, Yuan YG, Yu BL, Yang TJ and Cheng Y: Generation of human lactoferrin transgenic cloned goats using donor cells with dual markers and a modified selection procedure. Theriogenology. 78:1303–1311. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Ohkoshi K, Takahashi S, Koyama S, Akagi S, Adachi N, Furusawa T, Fujimoto J, Takeda K, Kubo M, Izaike Y and Tokunaga T: In vitro oocyte culture and somatic cell nuclear transfer used to produce a live-born cloned goat. Cloning Stem Cells. 5:109–115. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Wang M, Sun Z, Yu T, Ding F, Li L, Wang X, Fu M, Wang H, Huang J, Li N and Dai Y: Large-scale production of recombinant human lactoferrin from high-expression, marker-free transgenic cloned cows. Sci Rep. 7:107332017. View Article : Google Scholar : PubMed/NCBI

43 

Gordon JW: Studies of foreign genes transmitted through the germ lines of transgenic mice. J Exp Zool. 228:313–324. 1983. View Article : Google Scholar : PubMed/NCBI

44 

Hu J, Peng X, Schell TD, Budgeon LR, Cladel NM and Christensen ND: An hLA-A2.1-transgenic rabbit model to study immunity to papillomavirus infection. J Immunol. 177:8037–8045. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Yanagimachi R: Cloning: Experience from the mouse and other animals. Mol Cell Endocrinol. 187:241–248. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Oback B and Wells D: Donor cells for nuclear cloning: Many are called, but few are chosen. Cloning Stem Cells. 4:147–168. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Westhusin ME, Long CR, Shin T, Hill JR, Looney CR, Pryor JH and Piedrahita JA: Cloning to reproduce desired genotypes. Theriogenology. 55:35–49. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Robertson G, Garrick D, Wu W, Kearns M, Martin D and Whitelaw E: Position-dependent variegation of globin transgene expression in mice. Proc Natl Acad Sci USA. 92:5371–5375. 1995. View Article : Google Scholar : PubMed/NCBI

49 

Dobie KW, Lee M, Fantes JA, Graham E, Clark AJ, Springbett A, Lathe R and McClenaghan M: Variegated transgene expression in mouse mammary gland is determined by the transgene integration locus. Proc Natl Acad Sci USA. 93:6659–6664. 1996. View Article : Google Scholar : PubMed/NCBI

50 

Zhang J, Li L, Cai Y, Xu X, Chen J, Wu Y, Yu H, Yu G, Liu S, Zhang A, et al: Expression of active recombinant human lacto-ferrin in the milk of transgenic goats. Protein Expr Purif. 57:127–135. 2008. View Article : Google Scholar

51 

Tomita M, Wakabayashi H, Yamauchi K, Teraguchi S and Hayasawa H: Bovine lactoferrin and lactoferricin derived from milk: Production and applications. Biochem Cell Biol. 80:109–112. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 44 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang T, Yuan Y, Lu R, Xu S, Zhou M, Yuan T, Lu Y, Yan K and Cheng Y: The goat β‑casein/CMV chimeric promoter drives the expression of hLF in transgenic goats produced by cell transgene microinjection. Int J Mol Med 44: 2057-2064, 2019
APA
Zhang, T., Yuan, Y., Lu, R., Xu, S., Zhou, M., Yuan, T. ... Cheng, Y. (2019). The goat β‑casein/CMV chimeric promoter drives the expression of hLF in transgenic goats produced by cell transgene microinjection. International Journal of Molecular Medicine, 44, 2057-2064. https://doi.org/10.3892/ijmm.2019.4382
MLA
Zhang, T., Yuan, Y., Lu, R., Xu, S., Zhou, M., Yuan, T., Lu, Y., Yan, K., Cheng, Y."The goat β‑casein/CMV chimeric promoter drives the expression of hLF in transgenic goats produced by cell transgene microinjection". International Journal of Molecular Medicine 44.6 (2019): 2057-2064.
Chicago
Zhang, T., Yuan, Y., Lu, R., Xu, S., Zhou, M., Yuan, T., Lu, Y., Yan, K., Cheng, Y."The goat β‑casein/CMV chimeric promoter drives the expression of hLF in transgenic goats produced by cell transgene microinjection". International Journal of Molecular Medicine 44, no. 6 (2019): 2057-2064. https://doi.org/10.3892/ijmm.2019.4382