Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Scale-up production of and dietary supplementation with the recombinant antimicrobial peptide tilapia piscidin 4 to improve growth performance in Gallus gallus domesticus

  • Hsueh-Ming Tai ,

    Contributed equally to this work with: Hsueh-Ming Tai, Ming-Feng You

    Roles Formal analysis, Writing – original draft

    Affiliation Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Ilan, Taiwan

  • Ming-Feng You ,

    Contributed equally to this work with: Hsueh-Ming Tai, Ming-Feng You

    Roles Investigation, Methodology

    Affiliation Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Ilan, Taiwan

  • Chia-Hua Lin,

    Roles Formal analysis, Investigation

    Affiliation Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Ilan, Taiwan

  • Tsung-Yu Tsai,

    Roles Data curation, Formal analysis

    Affiliation Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Ilan, Taiwan

  • Chieh-Yu Pan ,

    Roles Formal analysis, Supervision, Validation

    panjade@nkust.edu.tw (CYP); zoocjy@gate.sinica.edu.tw (JYC)

    Affiliation Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan

  • Jyh-Yih Chen

    Roles Funding acquisition, Supervision, Writing – review & editing

    panjade@nkust.edu.tw (CYP); zoocjy@gate.sinica.edu.tw (JYC)

    Affiliation Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Ilan, Taiwan

Abstract

Antimicrobial peptides (AMPs) are short and positively charged peptides with broad-spectrum antimicrobial activities. AMPs have been investigated as potential antibiotic alternatives to improve growth performance and prevent pathogen infection in the poultry industry. The antimicrobial peptide tilapia piscidin 4 (TP4) was derived from Oreochromis niloticus, possesses antimicrobial activities and immunomodulatory properties, promotes intestinal health, and protects against pathogen infection. The codon-optimized sequence of TP4 was introduced into the pPICZαA vector and transformed into Pichia pastoris. Large-scale expression was induced following culture with methanol in a 500-liter fermenter. Freeze drying of fermented rTP4 broth and then rTP4 evaluation as a feed additive for Gallus gallus domesticus were performed. The in vitro antimicrobial activity of recombinant TP4 (rTP4) against gram-positive and gram-negative pathogens was evaluated. Evaluation of the effect of temperature on the antimicrobial activity of rTP4 showed its high stability at high temperatures. rTP4 significantly enhanced the phagocytic activity of macrophage cells, indicating that rTP4 has a remarkable ability to stimulate macrophages. rTP4 was used as a dietary supplement at 0.75, 1.5, 3.0, 6.0 and 12% in G. g. domesticus for five weeks, and growth performance, gut microbiota composition, and histology were assessed. The 3.0% rTP4 supplement group showed a significant increase in weight gain ratio and feed efficiency compared to those of the basal broiler diet group. Crude rTP4 was expressed by yeast to significantly promote growth efficiency and resistance against pathogens in G. g. domesticus, which could indicate its use as a suitable alternative to antibiotics as feed additives in the poultry industry.

1. Introduction

Antibiotics are commonly used to prevent disease and promote growth in agriculture. For poultry and livestock production, antibiotics are often used to treat clinical diseases and to control and prevent common infections. While antibiotics are used in animals for growth promotion or increased feed efficiency [1], the low concentration of antibiotics to which bacteria are exposed for a long period of time is problematic. A random mutation could cause some bacteria to become resistant to antibiotics. Drug-resistant microbes can survive during the antibiotic treatment period, and their population can be increased through natural selection. In recent decades, antibiotics have been used in poultry production as therapeutic agents to treat and control necrotic enteritis and respiratory diseases and to improve feed conversion and body weight gain [2]. These effects are due mainly to antibiotic treatment-induced gut microbiota alteration and microbiota modification in the gastrointestinal tract to provide an optimal microbiota for growth [3]. However, unlimited use of antibiotics will lead to reduced therapeutic effectiveness and generate many drug-resistant strains that may then have negative effects on animal and human health [4].

According to published data, different antibiotic‐resistant microbial strains have emerged, and the incidence of diseases caused by drug-resistant bacterial infection is increasing worldwide. There is an urgent need to study and introduce new types of effective, safe and natural compounds. Among these antimicrobial materials, AMPs have great potential as excellent alternatives to routine antibiotics used in the porcine and fowl industries [5]. Antimicrobial peptides possess extensive antimicrobial activities to inhibit gram-negative and gram-positive pathogens, fungi, viruses, and cancerous cells. Natural or artificial AMPs have been indicated to prevent bacterial colonization of cell surfaces, kill or inhibit microbes in biofilms, and destroy biofilm structures [6].

Most antimicrobial peptides are cationic peptides with conserved amphipathic molecular structure motifs that confer antibacterial properties [7]. Bacterial defense peptides were initially isolated from mammalian neutrophil granules, insect lymph and the skin of splendid leaf frogs [8] and demonstrated to have antimicrobial activities. Cationic defense peptides usually have few amino acids and are hydrophobic and hydrophilic charged [9]. The AMPs are then able to interact with the cytoplasmic membrane and rapidly destroy essential components inside the cells. Piscidins are histidine-enriched and linear antimicrobial peptides in the AMP family. The histidine content of piscidins and amphipathicity of peptides can cause different orientations of membrane insertion and permeability that very with pH value [10]. Piscidins isolated from teleost fish taxa consisting of 18 to 46 amino acids are expanded teleost-specific AMPs [11], which have a wide range of biological functions, including antibacterial, antiviral, antiparasite, antitumor, and immunomodulatory functions [1216].

The antimicrobial peptide piscidin was overexpressed in Escherichia coli, recovered from inclusion bodies and purified by Ni-NTA chromatography [1719]. However, in a prokaryotic expression system, expressed heterologous eukaryotic proteins may not be efficiently secreted into the broth and may fold incorrectly in large quantities. Yeast have been used to produce eukaryotic proteins in large amounts and active forms that are secreted into the medium. The antimicrobial peptide tilapia piscidin 4 (TP4) was derived and identified from Oreochromis niloticus and has activity against a broad range of microbial strains [2022]. In this study, a crude extract of a recombinant tilapia piscidin 4 (rTP4) culture was expressed by P. pastoris to obtain antimicrobial properties, which were tested against gram-positive and gram-negative pathogens of chickens and ducks. Simultaneously, the effect of dietary supplementation with the antimicrobial peptide rTP4 on the growth efficiency of G. g. domesticus was evaluated.

2. Methods

2.1 Microorganisms and animals

The bacterial strain Pichia pastoris X-33 (Invitrogen, CA, USA) was used as the TP4 expression host. Escherichia coli BL21 (DE3) (Stratagene, La Jolla, Calif., USA) was used as a DNA donor. DNA encoding TP4 with a His-tag was ligated into the pPICZαA (cat. no. V195-20, Invitrogen, CA, USA) expression vector. Procedures involving these animals adhered to the requirements of National Pingtung University of Science and Technology (NPUST) regarding their health and welfare and were approved by the Institutional Animal Care and Use Committee of NPUST (NPUST-107-026).

2.2. Construction and transformation of the expression recombinant plasmid and selection of positive transformants

To further increase the protein expression of recombinant TP4 in P. pastoris, the codon-optimized TP4 gene was designed to match P. pastoris expression by artificial gene synthesis from Omics Bio (Taipei, Taiwan). The nucleotide sequence products were confirmed by sequencing using primers, and then the identified plasmid was digested with EcoRI and XbaI. After enzyme digestion, the insert was ligated into the pPICZαA vector. The pPICZαA plasmid ligated with TP4 was digested with PmeI to linearize the vector and then transformed into P. pastoris X-33 by electroporation. The P. pastoris X-33 strain integrated with recombinant pPICZαA-TP4 DNA was grown in YPD broth at 30°C with 250 rpm agitation for 16 h. The resulting culture was inoculated into 30 mL of BMGY medium and incubated for 24 h at 30°C. The P. pastoris culture was then harvested by 10 min of centrifugation at 3,000 xg. The cell pellet was resuspended in 20 mM phosphate buffer, sonicated (400 cycles with 3 sec on/3 sec off) on ice by using a Sonicator 3000 system (HEAT Systems Inc., Farmingdale, NY) and then centrifuged at 5,000 xg for 20 min at 4°C. The recombinant protein in the pellet and supernatant was detected by SDS-PAGE and Western blotting using an anti-6x-His tag monoclonal antibody (Invitrogen, MA1-21315).

2.3. Scale-up production of recombinant protein in a 500-L fermenter

The stock was streaked on YPD agar plates with zeocin and incubated for two days at 30°C. BMGY with PTM4 broth and zeocin (100 μg/mL) was inoculated with a single colony and incubated in a shaker at 30°C and 150 rpm for two days. A second preculture consisting of 5% cultured broth was inoculated into YPD medium under the same conditions for one day. In this experiment, a 500-liter fermenter (BTF-C500L, Biotop Process & Equipment Inc, Nantou, Taiwan) was used with 350 liters of commercial medium. Fermentor cultivations were carried out by using basal salt medium and PTM1. During the fermentation period, the temperature was maintained at 30°C, and the pH was maintained and monitored at 6.0 with the addition of 14.5% ammonia and 0.1 N H2SO4. During the cultivation phase, the yeast were allowed to grow until the glycerol was exhausted after 36.5 h. Subsequently, the glycerol fed-batch phase consisted of feeding 50% (w/v) glycerol (containing 12 mL/L PTM) into the fermenter. After 47 h of cultivation, the methanol induction phase was performed by feeding 100% methanol (containing 16 mL/L PTM) for 24 h to induce rTP4 production. The rTP4-expressing yeast broth was harvested and freeze dried. Before rTP4 mixing with fodder preparation, rTP4 expression in the fermented culture and spray-dried powder was confirmed by Western blot analysis, and the rTP4 concentration was measured by comparing with different concentrations of synthetic TP4 peptide.

2.4. Antimicrobial activity of rTP4

The antimicrobial activity of rTP4 was measured in cultures of Escherichia coli (BCRC 10675), Pseudomonas aeruginosa (ATCC 19660), Staphylococcus aureus (BCRC 10780), and Riemerella anatipestifer (RA9, CFC27, CFC437, RA3, CFC363 and RA16). The microorganisms were collected from a single expanded colony and stored at -80°C. The microorganisms were inoculated in liquid broth and incubated at 37°C on a rotary shaker at 150 rpm overnight. The microorganisms were then diluted into fresh medium (1:1000) and incubated at 37°C on a rotary shaker. The bacterial cultures (104 CFU/ml) were mixed with 100 μl of an rTP4 solution and incubated at 37°C overnight. A negative control consisted of supernatant from P. pastoris transformed with vector alone, and ampicillin (2 mg/mL) was used as a positive control. After a 24-h incubation, the culture broth was measured at a wavelength of 600 nm. The antimicrobial activity of rTP4 was tested with an inhibition zone assay at different treatment temperatures [23]. Experiments were performed in duplicate and repeated independently three times.

2.5. G. g. domesticus maintenance and dietary treatment

A total of 119 male and 119 female 2-day-old G. g. domesticus were equally divided into seven groups for different dietary treatments, including basal diet, Spiraline-A (Juily Pharmaceutical Co., Ltd. San Sia, Taipei Hsien, Taiwan) and rTP4 groups (0.75, 1.5, 3.0, 6.0 and 12%). Thirty-four G. g. domesticus were housed in each cage with different treatment groups. The formulation of the basal diet and the composition analysis of the basal feed with additive in the early, middle and late stages are presented in S1 and S2 Tables. The chickens had ad libitum access to water and fodder for the duration of the trial from 3 to 36 days of age. The animals were kept in a 12:12 light-dark cycle at 18.4–31.2°C in a cage with a constant environment. The rTP4 feed, fecal sludge and wastewater were not discarded directly into the environment. Statistical analysis was performed with Student’s t-test when comparing two groups. Multiple group comparisons were performed by ANOVA in GraphPad Prism 9. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

2.6. The analysis of the phagocytic activity and cell size and complexity assessments

The effect of rTP4 on phagocytic activity was studied with the use of GFP-tagged E. coli bacteria using flow cytometry. The phagocytosis of GFP-E. coli (ATCC 25922GFP™) was analyzed by monitoring the mean GFP fluorescence intensity of engulfed bacteria. RAW264.7 cells were preincubated with rTP4 in 10% FBS/DMEM with gentle shaking for 24 h at 37°C. The rTP4-induced RAW264.7 macrophage cells were allowed to uptake and become infected with GFP-labeled E. coli (107 cfu/mL) at an infective concentration of 1:20 (MOI) for an additional 4 h. The change in phagocytic activity was assessed according to the change in fluorescence intensity. After rTP4 stimulation, cells were removed from the culture dishes using a cell scraper and then centrifuged and washed with PBS. The cells were resuspended in a PBS solution. Flow cytometer (Beckman Coulter) analysis was performed using forward (FSC: cell size) and side scatter (SSC: cell complexity) parameters.

2.7. Physiological analysis and histological procedures

During the experimental period, the feed intake, body weight and survival rate were measured. Weight gain (WG), protein efficiency ratio (PER), feed efficiency (FE), and survival percentage were calculated. After 36 days of the feeding trial, G. g. domesticus were euthanized, and blood serum samples were collected after centrifugation (3,000 ×g for 10 min) and stored at -80°C for analysis. The specimens were fixed in 4% formaldehyde in 0.1 M sodium phosphate buffer (PBS, pH 7.2) at room temperature. Subsequently, they were rinsed in PBS and dehydrated in graded ethanol. Tissue sections were deparaffinized in xylene and hydrated using serial percentages of alcohol. The sections were stained with hematoxylin for 10 min and eosin for 5 min.

2.8. Gut microbiota analysis

The duodenum stool of G. g. domesticus fed with the basal, Spiraline-A, or rTP4 diet was dissected for genomic DNA extraction using an innuPREP stool DNA kit (Jena AG, Jena, Germany). The DNA concentration was measured by using a NanoDrop ND1000 spectrophotometer (NanoDrop® Technologies), and DNA was stored at -20°C until use. The V4 region of the 16S rRNA gene was amplified with the forward PCR primer 515F and reverse primer 806R. All samples were amplified on a Veriti 96-well Thermal Cycler (Applied Biosystems, Foster City, USA). PCR amplicon sequencing was performed by the Biotools Microbiome Research Center (New Taipei City, Taiwan). The sequences were then processed using QIIME software [24]. QIIME can perform taxonomic assignment at the genus level on the sequencing from the Biotools Microbiome Research Center.

3. Results

3.1. Construction of a TP4 expression pPICZαA plasmid

DNA encoding TP4 with a 6xHis tag was inserted into the pPICZαA expression vector to obtain the α-Factor-TP4-6xHis expression cassette (Fig 1). The pPICZαA plasmid ligated with TP4 was digested with PmeI within the AOX1 region to linearize the pPICZαA vector, and then TP4 genes were transformed into the Pichia pastoris X-33 strain. Colonies were screened by plating on YPDS plates containing different concentrations of zeocin. Transformed P. pastoris X-33 colonies integrated with recombinant pPICZαA-TP4 DNA were then selected by colony hybridization by using an anti-6xHis tag antibody, and high-expression clones were picked for subsequent experiments (previous data). The transformants were confirmed by colony PCR analysis with AOX1 primers (previous data).

thumbnail
Fig 1. Scale-up expression of rTP4 in P. pastoris in a 500-liter fermenter.

(A) The dissolved oxygen (%), pH, RPM and temperature (°C) of broth during incubation of a P. pastoris X-33 transformant in BSM. (B) Western blot analysis profiles of recombinant rTP4 expression. M: Molecular weight marker; sTP4: Synthetic TP4 with concentrations of 10, 5, 2.5, 1.25 and 0.635 ng/μL; rTP4: Recombinant TP4.

https://doi.org/10.1371/journal.pone.0253661.g001

3.2. Construction of the TP4 expression pPICZαA plasmid and large-scale expression of recombinant TP4 in a 500-L fermenter

DNA encoding TP4 with a 6xHis tag was inserted into the pPICZαA expression vector to obtain the α-Factor-TP4-6xHis expression cassette, which was described in our previous report [25]. Recombinant TP4 was expressed in the methylotrophic yeast P. pastoris under the control of the AOX1 promoter and secreted into the broth using the α-factor pre-pro sequence with the STE13 gene for dipeptidyl aminopeptidase A (Fig 1). After the successful production of the rTP4 transformant, the cells were cultured in a 5-liter bench-top fermenter. The parameters of dissolved oxygen (DO), RPM, pH and agitation were optimized for rTP4 production, and the effect of induction time of rTP4 expression was evaluated (previous data). We enlarged the scale from a 5 to 500 L fermenter with a two-stage fed-batch cultivation. Five percent of a P. pastoris culture was inoculated into 350 L of BSM. The cultivation process is shown in Fig 2A. During 36.5 h of cultivation, glycerol content decreased with time, and we began to feed glycerol into the fermentation. When the P. pastoris transformant was growing to stationary phase, the highest viable count reached approximately 8.3×1010 CFU/mL at 48 h of cultivation. At the same time, methanol inductions were carried out in which the viable count and wet weight of yeast showed a sharp decrease (Fig 2A). After 65 h of cultivation, the fermented broth was harvested and then freeze dried to powder. The rTP4 powder was confirmed by Western blot analysis, and the rTP4 concentration was measured at 1.07 mg/g by comparison with different concentrations of synthetic TP4 peptide (Fig 2B). rTP4 powder was used as a feed additive for dietary supplementation in G. g. domesticus.

thumbnail
Fig 2. The effects of rTP4 on gram-negative and gram-positive strains by coincubation assay.

After incubation, the culture medium was measured at a wavelength of 600 nm. A low OD600 value indicates that bacterial growth was inhibited. Vector control: Empty pPICZαA vector. TP4: Protein expressed by the pPICZαA-TP4 vector.

https://doi.org/10.1371/journal.pone.0253661.g002

3.3. Antimicrobial activity of rTP4 expression in P. pastoris

To evaluate the antimicrobial activity of the rTP4 protein, the culture was induced with methanol and mixed with equal volumes of diluted bacterial cultures of gram-positive and gram-negative pathogens. After 16 h of incubation, the culture medium was measured at a wavelength of 600 nm. Compared to P. pastoris with a vector control, which did not inhibit pathogen growth, rTP4 showed high activity to inhibit pathogen growth (Fig 3). Meanwhile, the Kirby-Bauer disk diffusion method was used for antimicrobial susceptibility testing of rTP4, and the results indicated that rTP4 had a larger diameter and a more visible zone against the tested strains than the control treatment. The antimicrobial peptide was most effective against the pathogens R. anatipestifer (RA3) and R. anatipestifer (CFC27) (Table 1A). Furthermore, a study of the thermal stability of rTP4 was performed against S. aureus (BCRC 10780), P. aeruginosa (ATCC 19660) and E. coli (BCRC 10675). The thermal stability of the antimicrobial peptide was tested at 40, 60, 80 or 100°C for 5 min, and the selected pathogens were detected by the agar disk diffusion method. The negative control group was P. pastoris with the empty vector. The antimicrobial peptide rTP4 showed good temperature stability at high temperature, inhibiting S. aureus and E. coli growth. The P. aeruginosa strain was inhibited by the antibacterial activity of rTP4 with increasing temperature (Table 1B). The results showed that heat treatment of rTP4 did not significantly affect the antimicrobial ability at temperatures up to 100°C.

thumbnail
Fig 3. Flow cytometry phagocytosis assay.

Flow cytometry analysis using cell size forward-scattered light (FSC) and side-scattered light (SSC) parameters was performed. (A) RAW264.7 cells alone, (B) RAW264.7 cells coincubated with E. coli, and (C) RAW264.7 cells treated with rTP4 and coincubated with E. coli. (D) The mean fluorescence intensity (MFI) of RAW264.7 cells was analyzed, and the asterisks indicate significant differences.

https://doi.org/10.1371/journal.pone.0253661.g003

thumbnail
Table 1. The antimicrobial activity and effect of temperature of rTP4.

The rTP4 was expressed by P. pastoris X-33 and supernatant were collected after induction and applied to disk diffusion assay. After incubation, the inhibition zone diameters were measured on the culture plate. Representative radial diffusion assays are shown. (A) The antimicrobial activity of rTP4. (B) The effect of temperature on rTP4 antimicrobial activity.

https://doi.org/10.1371/journal.pone.0253661.t001

3.4. Effects of rTP4 on cell size and complexity in a phagocytic activity assay

To evaluate rTP4 in vitro stimulation of macrophage differentiation, the RAW264.7 cell line was treated with rTP4 for 24 h. Flow cytometry analysis using cell size forward-scattered light (FSC) and side-scattered light (SSC) parameters was performed. The results showed that the cell size and complexity after GFP-E. coli phagocytosis by RAW264.7 cells without rTP4 treatment were similar to those of RAW264.7 cells alone (Fig 3A and 3B). After rTP4 stimulation, the immunogenicity increased with molecular size and complexity in the macrophage differentiation assay (Fig 3C). In addition, increased cell complexity may be associated with increased storage or secretory vesicles, such as lysosomes. The mean fluorescent intensity (MFI) showing phagocytosis of GFP-E. coli in RAW264.7 cells after treatment with rTP4 was higher than that in the other groups (Fig 3D).

3.5. rTP4 supplementation improves growth performance and histology

After 36 days of the experimental period, the growth performance of G. g. domesticus in percent weight gain (WG) and feed efficiency (FE) in 3.0% rTP4-fed animals was significantly higher than that in the basal diet group animals and similar to that in the Spiraline-A group animals (Table 2). The protein efficiency ratios of G. g. domesticus in the 0.75% and 1.5% rTP4-fed animals were higher than those in the animals in other groups (Fig 4). The survival rate of animals fed 1.5% rTP4 was lower than that of the animals in other groups, which was caused by one chicken dying from disease and two chickens dying after pecking an infected carcass. Morphological measurements of the heart, liver, spleen, lung, kidney and intestine are given in Fig 5. Histological examination of tissues showed white blood cell aggregates in the coronary vein of the heart in the antibiotic group (Fig 5A, black arrow). This phenomenon was not found in the coronary veins of the 3% rTP4 group or basal diet group. Microscopic examination of tissue sections, including those of the heart, liver, spleen, lung, kidney and intestine, in the three groups was normal, and no other symptoms were observed. Microbial profiles of the duodenum in chickens with different treatments were determined using high-throughput sequencing of the bacterial 16S rRNA gene. The results showed that the duodenum was predominantly populated by Peptostreptococcaceae and Erysipelotrichaceae. Reduced total viable counts of Ruminococcaceae and Lachnospiraceae were also observed after feeding with rTP4 (Fig 6).

thumbnail
Fig 4. Effect on feed parameter analysis.

(A) Weight for day 2 and (B) day 36 analysis between groups. (C) weight gain, (D) FE (feed efficiency), and (E) PER (protein efficiency ratio) of G. g. domesticus fed diets containing rTP4 (0.75, 1.5, 3.0, 6.0 or 12%) spray-dried powder for 5 weeks. Each bar represents the mean value from three determinations, with the standard error (SE). Data (mean ± SE) with star marker represent differ significantly (P<0.05) among treatments.

https://doi.org/10.1371/journal.pone.0253661.g004

thumbnail
Fig 5. Histological analysis of tissue sections.

Representative photomicrographs of tissue sections stained with H&E. A: Spiraline-A group (No. 50); B: 3% rTP4 group (No. 99); C: Basal diet group (No. 90).

https://doi.org/10.1371/journal.pone.0253661.g005

thumbnail
Fig 6. Effect of orally administered 3% rTP4, Spiraline-A and basal diet on the intestinal microflora in G. g. domesticus.

Control: Basal diet group; AMP: 3% rTP4 group; antibio: Spiraline-A group.

https://doi.org/10.1371/journal.pone.0253661.g006

thumbnail
Table 2. Effect of weight gain ratio, feed efficiency (FE), protein efficiency ratio (PER) and survival rate of G. g. domesticus fed with diets containing rTP4 (0.75, 1.5, 3.0, 6.0 and 12%) spray-dried powder for 5 weeks1.

https://doi.org/10.1371/journal.pone.0253661.t002

4. Discussion

Antibiotics are widely used to prevent pathogen infection and promote growth in poultry and livestock production [26]. However, many reports indicate that long-term use of low-dose antibiotics may lead to resistant microorganisms. For various reasons, the use of antibiotics has been banned or restricted for growth promotion in the poultry industry in many countries. Consequently, there is a high demand for antibiotic alternatives in the livestock industry. Compared with traditional antibiotics, AMPs have been shown to have multiple biological functions in addition to antimicrobial activities, including antibacterial, antiviral, antiprotozoan and antifungal activities [27]. In other respects, antimicrobial peptides protect fish against infections caused by the fish pathogen Vibrio anguillarum and provide a concept of transgenic fish with antimicrobial peptides. Antimicrobial peptides may decrease fish bacterial disease [28]. Antimicrobial peptides (AMPs) also stimulate the innate immune system via immunomodulatory functions and modulate the intestinal microflora in animals and humans [5]. In addition, AMPs had positive effects on intestinal morphology, nutrient digestibility, the gut microbiota, and growth performance in animals. These AMPs have a strong potential for application as a feed additive as alternatives to antibiotics in swine and poultry production. To study the role of antimicrobial peptides and mass production as feed supplements by use in the livestock industry, we performed this research. Yeast expression systems are ideally suited for large-scale production and cost-effective expression of functional AMP proteins. According to the codon usage preference of P. pastoris, the mature TP4 cDNA was modified, synthesized and cloned into the pPICZαA vector for P. pastoris X-33 expression.

There are many antimicrobial peptides that have been expressed in P. pastoris and possess natural antibacterial activities, such as ABP-dHC-cecropin A human neutrophil peptide 1 (HNP1) and histidalin [2931]. In this study, we successfully produced recombinant TP4 on a 500-liter scale and freeze dried it to use as a feed additive for dietary supplementation in broilers. In the second cultivation, the viable count declined quickly from 8.3×1010 to 6.5×1010 CFU/mL, and the wet weight declined from 275 to 166 g/L during 49 to 58 hours of cultivation. This effect is presumably caused by methanol-induced antimicrobial peptide production to inhibit host cell growth [32]. According to previous research, the antimicrobial peptide HKPLP derived from Hippocampus kuda Bleeker showed good heat stability in thermal stability analysis [33]. After high-temperature treatment, rTP4 expressed by P. pastoris was found to be very stable in vitro and still retained full antimicrobial activity against S. aureus (BCRC 10780) and E. coli (BCRC 10675). The thermal stability of rTP4 may be related to its high arginine content (24%, 5 arginine in 25 total residues) [34]. In a previous report, increased arginine content enhanced peptide insertion into the membrane and significantly increased antimicrobial activity [35]. In addition, the antibacterial activity of AMPs can be enhanced by modifying the structural properties of natural peptides [3638]. In a similar report, the crude antimicrobial extract of Bacillus velezensis strains isolated from stingless bee products showed bactericidal activity against methicillin-resistant Staphylococcus aureus and was stable at various temperatures (40–80°C) [39]. Furthermore, Plantaricin LD1 produced by Lactobacillus plantarum LD1 showed stability at high temperatures (100°C for 20 min and 121°C for 15 min under 15 psi) and inhibited bacteria such as Staphylococcus aureus, urogenic Escherichia coli, Pseudomonas aeruginosa, Salmonella typhi, Shigella flexneri and Vibrio sp [40]. Additionally, cecropinXJ expressed in Escherichia coli retained high stability against Staphylococcus aureus over a temperature range from 4 to 100°C [41].

These results show that compared to the basal diet, fodder supplementation with crude rTP4 can improve the growth of G. g. domesticus. Regarding the antibiotic group, the 1.5% rTP4 and 3.0% rTP4 groups were similar, indicating that the antimicrobial peptide rTP4 can be recommended as an alternative to antibiotics to promote growth performance in chickens. According to this study, the FCR of the basal diet group was around 2.0, the Spiraline-A (commercial) group was around 1.88, and the 3% rTP4-fed group was around 1.85. In our experience, typical FCRs for animals raised using commercial feeds and intensive production methods are as follows: beef cattle: 6.0–10.0, pigs: 2.7–5.0, farmed fish and shrimp: 1.0–2.4, and chickens: 1.7–2.0. The FCRs in this study range from 1.85 to 2.0, which is within the normal range for chickens. In previous research, similar to an antibiotic, the antimicrobial peptide cLF36 improved the immune cells, intestinal morphology, intestinal microbiome, junctional proteins, and growth performance in E. coli-challenged broilers [42]. Pigs fed AMP‐P5 at 60 mg kg-1 in dietary supplementation showed enhanced growth, inhibited coliform growth and increased nutrient digestion [43]. Tilapia piscidin, a marine antimicrobial peptide, has antimicrobial and immune regulatory activities that make it a potentially valuable feed supplement in aquaculture. Dietary supplementation with tilapia piscidin could enhance intestinal health, antioxidant activity, and the immune response. Furthermore, it confers protection against pathogen infections in Nile tilapia [44]. Morphological measurements of the heart, liver, spleen, lung, kidney and intestine showed white blood cell aggregates in the coronary vein of the heart in the broilers of the antibiotic group. The phenomenon of white blood cell aggregates is associated with infection or inflammation in the body. The Spiraline-A antibiotic group contained streptomycin sulfate. Streptomycin is administered by intramuscular injections in humans. Repeated injections of streptomycin can cause pain and inflammation [45]. Some reports have shown that the cecum mucosa of streptomycin-treated mice has mild inflammatory infiltration [46]. In the gut microbiota community richness analysis, the total viable counts of the Ruminococcaceae and Lachnospiraceae were reduced. Peptostreptococcaceae and Erysipelotrichaceae were enriched in the duodenum in the TP4 group compared with in the antibiotic and basal diet groups. A report showed an increase in Lachnospiraceae in WIRS mice, which is associated with human diseases, such as ulcerative colitis, celiac disease and Crohn’s disease [47].

Conclusions

This study revealed that recombinant TP4 expressed by yeast has high antimicrobial activity and thermal stability. Therefore, rTP4 is available for feed application, as the antimicrobial peptide can withstand the high temperature used in pelleted feed procedure and still have strong antibacterial activity. These results showed that the antimicrobial peptide tilapia piscidin 4 can be used as a feed additive for the livestock industry.

Supporting information

S1 Fig. Plasmid map of the pPICZαA-TP4 vector.

https://doi.org/10.1371/journal.pone.0253661.s001

(PPT)

S1 Table. Proximate analysis of basal feed with additive compositions at the early, middle and late stages.

https://doi.org/10.1371/journal.pone.0253661.s003

(DOC)

S2 Table. Formulation of the basal diet at the early, middle and late stages.

https://doi.org/10.1371/journal.pone.0253661.s004

(DOC)

Acknowledgments

We would like to thank American Journal Experts for language editing.

References

  1. 1. Chattopadhyay M. K., Use of antibiotics as feed additives: a burning question. Front Microbiol 2014, 5. pmid:24523715
  2. 2. Diarra M. S.; Malouin F., Antibiotics in Canadian poultry productions and anticipated alternatives. Front Microbiol 2014, 5. pmid:24523715
  3. 3. Singh P.; Karimi A.; Devendra K.; Waldroup P. W.; Cho K. K.; Kwon Y. M., Influence of penicillin on microbial diversity of the cecal microbiota in broiler chickens. Poultry Sci 2013, 92 (1), 272–276. pmid:23243258
  4. 4. Mehdi Y.; Letourneau-Montminy M. P.; Gaucher M. L.; Chorfi Y.; Suresh G.; Rouissi T.; et al, Use of antibiotics in broiler production: global impacts and alternatives. Anim Nutr 2018, 4 (2), 170–178. pmid:30140756
  5. 5. Wang S.; Zeng X. F.; Yang Q.; Qiao S. Y., Antimicrobial peptides as potential alternatives to antibiotics in food animal industry. Int J Mol Sci 2016, 17 (5). pmid:27153059
  6. 6. Yasir M.; Willcox M. D. P.; Dutta D., Action of antimicrobial peptides against bacterial biofilms. Materials 2018, 11 (12). pmid:30563067
  7. 7. Hancock R. E. W.; Sahl H. G., Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol 2006, 24 (12), 1551–1557. pmid:17160061
  8. 8. Proano-Bolanos C.; Mei Z. A.; Wang L.; Coloma L. A.; Chen T. B.; Shaw C., Peptidomic approach identifies cruzioseptins, a new family of potent antimicrobial peptides in the splendid leaf frog, Cruziohyla calcarifer. J Proteomics 2016, 146, 1–13. pmid:27321580
  9. 9. Mookherjee N.; Hancock R. E. W., Cationic host defence peptides: Innate immune regulatory peptides as a novel approach for treating infections. Cell Mol Life Sci 2007, 64 (7–8), 922–933. pmid:17310278
  10. 10. Mihailescu M.; Sorci M.; Seckute J.; Silin V. I.; Hammer J.; Perrin B. S.; et al, Structure and function in antimicrobial piscidins: histidine position, directionality of membrane insertion, and pH-dependent permeabilization. J Am Chem Soc 2019, 141 (25), 9837–9853. pmid:31144503
  11. 11. Salger S. A.; Cassady K. R.; Reading B. J.; Noga E. J., A Diverse family of host-defense peptides (piscidins) exhibit specialized anti-bacterial and anti-protozoal activities in fishes. Plos One 2016, 11 (8). pmid:27552222
  12. 12. Chen W. F.; Huang S. Y.; Liao C. Y.; Sung C. S.; Chen J. Y.; Wen Z. H., The use of the antimicrobial peptide piscidin (PCD)-1 as a novel anti-nociceptive agent. Biomaterials 2015, 53, 1–11. pmid:25890701
  13. 13. Peng K. C.; Lee S. H.; Hour A. L.; Pan C. Y.; Lee L. H.; Chen J. Y., Five different piscidins from nile tilapia, Oreochromis niloticus: analysis of their expressions and biological functions. Plos One 2012, 7 (11). pmid:23226256
  14. 14. Ting C. H.; Lee K. Y.; Wu S. M.; Feng P. H.; Chan Y. F.; Chen Y. C.; et al FOSB-PCDHB13 axis disrupts the microtubule network in non-small cell lung cancer. Cancers 2019, 11 (1). pmid:30658436
  15. 15. Ting C. H.; Chen Y. C.; Wu C. J.; Chen J. Y., Targeting FOSB with a cationic antimicrobial peptide, TP4, for treatment of triple-negative breast cancer. Oncotarget 2016, 7 (26), 40329–40347. pmid:27248170
  16. 16. Colorni A.; Ullal A.; Heinisch G.; Noga E. J., Activity of the antimicrobial polypeptide piscidin 2 against fish ectoparasites. J Fish Dis 2008, 31 (6), 423–432. pmid:18471098
  17. 17. Pan Y.; Zheng L. B.; Mao Y.; Wang J.; Lin L. S.; Su Y. Q.; et al, The antibacterial activity and mechanism analysis of piscidin 5 like from Larimichthys crocea. Dev Comp Immunol 2019, 92, 43–49. pmid:30359623
  18. 18. Moon W. J.; Hwang D. K.; Park E. J.; Kim Y. M.; Chae Y. K., Recombinant expression, isotope labeling, refolding, and purification of an antimicrobial peptide, piscidin. Protein Expres Purif 2007, 51 (2), 141–146.
  19. 19. Chen W.; Cotten M. L., Expression, purification, and micelle reconstitution of antimicrobial piscidin 1 and piscidin 3 for NMR studies. Protein Expres Purif 2014, 102, 63–68.
  20. 20. Wang S. H.; Wang C. F.; Chang T. W.; Wang Y. J.; Liao Y. D., Oligomerization and insertion of antimicrobial peptide TP4 on bacterial membrane and membrane-mimicking surfactant sarkosyl. Plos One 2019, 14 (5). pmid:31083701
  21. 21. Pan C. Y.; Tsai T. Y.; Su B. C.; Hui C. F.; Chen J. Y., Study of the Antimicrobial Activity of Tilapia Piscidin 3 (TP3) and TP4 and Their Effects on Immune Functions in Hybrid Tilapia (Oreochromis spp.). Plos One 2017, 12 (1).
  22. 22. Narayana J. L.; Huang H. N.; Wu C. J.; Chen J. Y., Efficacy of the antimicrobial peptide TP4 against Helicobacter pylori infection: in vitro membrane perturbation via micellization and in vivo suppression of host immune responses in a mouse model. Oncotarget 2015, 6 (15), 12936–12954. pmid:26002554
  23. 23. Peng Z.; Wang A.; Feng Q.; Wang Z.; Ivanova I. V.; He X.; et al, High-level expression, purification and characterisation of porcine beta-defensin 2 in Pichia pastoris and its potential as a cost-efficient growth promoter in porcine feed. Appl Microbiol Biotechnol 2014, 98 (12), 5487–97. pmid:24515729
  24. 24. Caporaso J. G.; Kuczynski J.; Stombaugh J.; Bittinger K.; Bushman F. D.; Costello E. K.; et al QIIME allows analysis of high-throughput community sequencing data. Nat Methods 2010, 7 (5), 335–6. pmid:20383131
  25. 25. Cromwell G. L., Why and how antibiotics are used in swine production. Anim Biotechnol 2002, 13 (1), 7–27. pmid:12212945
  26. 26. Zanetti M., Cathelicidins, multifunctional peptides of the innate immunity. J Leukocyte Biol 2004, 75 (1), 39–48. pmid:12960280
  27. 27. Jia X.; Patrzykat A.; Devlin R. H.; Ackerman P. A.; Iwama G. K.; Hancock R. E. W., Antimicrobial peptides protect coho salmon from Vibrio anguillarum infections. Appl Environ Microb 2000, 66 (5), 1928–1932. pmid:10788362
  28. 28. Sang M.; Wei H.; Zhang J.; Wei Z.; Wu X.; Chen Y.; et al, Expression and characterization of the antimicrobial peptide ABP-dHC-cecropin A in the methylotrophic yeast Pichia pastoris. Protein Expr Purif 2017, 140, 44–51. pmid:28827052
  29. 29. Zhang X. L.; Jiang A. M.; Qi B. H.; Yu H.; Xiong Y. Y.; Zhou G. L.; et al, Secretion expression of human neutrophil peptide 1 (HNP1) in Pichia pastoris and its functional analysis against antibiotic-resistant Helicobacter pylori. Appl Microbiol Biot 2018, 102 (11), 4817–4827.
  30. 30. Meng D. M.; Li W. J.; Shi L. Y.; Lv Y. J.; Sun X. Q.; Hu J. C.; et al, Expression, purification and characterization of a recombinant antimicrobial peptide Hispidalin in Pichia pastoris. Protein Expr Purif 2019, 160, 19–27. pmid:30904445
  31. 31. Li Y. F., Recombinant production of antimicrobial peptides in Escherichia coli: A review. Protein Expres Purif 2011, 80 (2), 260–267. pmid:21843642
  32. 32. Sun D.; Wu S.; Jing C.; Zhang N.; Liang D.; Xu A., Identification, synthesis and characterization of a novel antimicrobial peptide HKPLP derived from Hippocampus kuda Bleeker. J Antibiot (Tokyo) 2012, 65 (3), 117–121. pmid:22252202
  33. 33. Stensvag K.; Haug T.; Sperstad S. V.; Rekdal O.; Indrevoll B.; Styrvold O. B., Arasin 1, a proline-arginine-rich antimicrobial peptide isolated from the spider crab, Hyas araneus. Dev Comp Immunol 2008, 32 (3), 275–285. pmid:17658600
  34. 34. Cutrona K. J.; Kaufman B. A.; Figueroa D. M.; Elmore D. E., Role of arginine and lysine in the antimicrobial mechanism of histone-derived antimicrobial peptides. Febs Lett 2015, 589 (24), 3915–3920. pmid:26555191
  35. 35. Maloy W. L.; Kari U. P., Structure-activity studies on magainins and other host-defense peptides. Biopolymers 1995, 37 (2), 105–122. pmid:7893944
  36. 36. Javadpour M. M.; Juban M. M.; Lo W. C. J.; Bishop S. M.; Alberty J. B.; Cowell S. M.; et al, De novo antimicrobial peptides with low mammalian cell toxicity. J Med Chem 1996, 39 (16), 3107–3113. pmid:8759631
  37. 37. Dathe M.; Nikolenko H.; Meyer J.; Beyermann M.; Bienert M., Optimization of the antimicrobial activity of magainin peptides by modification of charge. Febs Lett 2001, 501 (2–3), 146–150. pmid:11470274
  38. 38. Daneshmand A.; Kermanshahi H.; Sekhavati M. H.; Javadmanesh A.; Ahmadian M., Antimicrobial peptide, cLF36, affects performance and intestinal morphology, microflora, junctional proteins, and immune cells in broilers challenged with E. coli. Sci Rep-Uk 2019, 9.
  39. 39. Baharudin M.M.A.; Ngalimat M.S.; Mohd Shariff F.; Balia Yusof Z.N.; Karim M.; Baharum S.N.; et al, Antimicrobial activities of Bacillus velezensis strains isolated from stingless bee products against methicillin-resistant Staphylococcus aureus. PLoS One 2021, 16(5), e0251514. pmid:33974665
  40. 40. Gupta A.; Tiwari S.K., Plantaricin LD1: a bacteriocin produced by food isolate of Lactobacillus plantarum LD1. Appl Biochem Biotechnol 2014, 172(7), 3354–3362. pmid:24522411
  41. 41. Xia L.; Zhang F.; Liu Z.; Ma J.; Yang J., Expression and characterization of cecropinXJ, a bioactive antimicrobial peptide from Bombyx mori (Bombycidae, Lepidoptera) in Escherichia coli. Exp Ther Med 2013, 5(6), 1745–1751. pmid:23837066
  42. 42. Yoon J. H.; Ingale S. L.; Kim J. S.; Kim K. H.; Lohakare J.; Park Y. K.; et al, Effects of dietary supplementation with antimicrobial peptide-P5 on growth performance, apparent total tract digestibility, faecal and intestinal microflora and intestinal morphology of weanling pigs. J Sci Food Agr 2013, 93(3), 587–592. pmid:22903784
  43. 43. Zahran E.; Risha E.; Elbahnaswy S.; Mahgoub H. A.; Abd El-Moaty A., Tilapia piscidin 4 (TP4) enhances immune response, antioxidant activity, intestinal health and protection against Streptococcus iniae infection in Nile tilapia. Aquaculture 2019, 513.
  44. 44. Tanoira R. P.; Sanchez-Patan F.; Giron A. J.; Roblas R. F.; Esteban J.; Guerrero M. L. F., Tolerance and safety of intravenous streptomycin therapy in patients with tuberculosis. Infection 2014, 42 (3), 597–598. pmid:24748409
  45. 45. Spees A. M.; Wangdi T.; Lopez C. A.; Kingsbury D. D.; Xavier M. N.; Winter S. E.; et al, Streptomycin-induced inflammation enhances Escherichia coli gut colonization through nitrate respiration. Mbio 2013, 4 (4). pmid:23820397
  46. 46. Zhu C.; Song W.; Tao Z.; Liu H.; Zhang S.; Xu W.; et al, Analysis of microbial diversity and composition in small intestine during different development times in ducks. Poult Sci 2020, 99 (2), 1096–1106. pmid:32029146
  47. 47. Li S. Y.; Wang Z. L.; Yang Y.; Yang S.; Yao C. C.; Liu K. Y.; et al, Lachnospiraceae shift in the microbial community of mice faecal sample effects on water immersion restraint stress. Amb Express 2017, 7. pmid:28050848