Skip to main content
  • Research article
  • Open access
  • Published:

Development of a novel chimeric lysin to combine parental phage lysin and cefquinome for preventing sow endometritis after artificial insemination

Abstract

Sow endometritis is usually caused by multiple species of pathogenic bacteria. Numerous isolates from endometritis patients have developed antimicrobial resistance. Thus, novel antibacterial agents and strategies to combat endometritis are needed. A total of 526 bacteria, including Staphylococcus spp. (26.3%), Streptococcus spp. (12.3%), E. coli (28.9%), Enterococcus spp. (20.1%), Proteus spp. (9.5%), and Corynebacterium spp. (2.8%), were isolated from sows with endometritis. We constructed a novel chimeric lysin, ClyL, which is composed of a cysteine- and histidine-dependent amidohydrolase/peptidase (CHAP) catalytic domain from the phage lysin LysGH15 and a cell wall-binding domain (CBD) from the prophage lysin Lys0859. The activities of ClyL and Lys0859 were most pronounced for the Staphylococcus and Streptococcus strains isolated from sow endometritis and bovine mastitis, respectively. ClyL and Lys0859 were combined to create a phage lysin cocktail, which demonstrated a synergistic effect against the coinfection of Staphylococcus and Streptococcus in vitro and in vivo. Furthermore, the combination of phage lysin cocktail and cefquinome had a synergistic bactericidal effect on boar semen that did not influence the activity of sperm. Remarkably, the incidence rate of sow endometritis was 0% (0/7) when the combination of phage lysin cocktail and cefquinome was used in semen via artificial insemination compared with 50% (3/6) when PBS was administered. Overall, the administration of a phage lysin cocktail and cefquinome in semen via artificial insemination is a promising novel strategy to prevent sow endometritis after artificial insemination.

Introduction

Endometritis, an inflammatory disease, seriously affects the reproductive performance of sows (including the sensitivity of piglets to infection, growth, and mortality) and leads to enormous economic losses [1,2,3]. It is caused predominantly by common bacteria (including Staphylococcus aureus, Streptococcus spp., and Escherichia coli), viruses (including Japanese encephalitis, porcine reproductive, and respiratory syndrome virus), and parasites (e.g., Trichomonas) [3,4,5,6]. Ye et al. isolated a total of 556 bacterial strains from 200 sows across 11 farms located in Guangdong and Jiangsu Provinces, China. S. aureus was the most prevalent isolate (34%, n = 188), followed by Streptococcus (32%, n = 181), E. coli (19%, n = 104) and other bacilli (15%, n = 83) [3]. A significant proportion of sow endometritis cases (83%, n = 207) were identified as mixed infections, complicating clinical treatment efforts [3]. The microbiota results of birth canal secretions and fresh feces from healthy sows and sows with endometritis revealed that the abundances of Escherichia–Shigella and Streptococcus in sows suffering from endometritis were significantly greater than those in healthy sows [1]. Bacterial-induced endometritis has a significant effect on the reproductive capabilities of both animals and humans, contributing to the death of 6.6% of breeding sows on large-scale farms [7,8,9]. Cow endometritis can lead to embryo transfer failure [8]. Antibiotic therapy can significantly improve the rate of successful pregnancies in women with CE undergoing IVF [10]. Similarly, the pregnancy rate of the mare was significantly improved by intrauterine infusion of broad-spectrum antibiotics compared with that of the untreated group (P < 0.01) [11]. However, isolates from animals and humans with endometritis commonly exhibit antimicrobial resistance (AMR) [2, 12, 13], and AMR has become a global health concern that has been further exacerbated by the slow development of novel antibiotics. Previous research has shown that lysostaphin can be used to treat clinical sow endometritis via intravaginal administration [3]. Following treatment with lysostaphin at a dosage of 400 U, the average cure rate of lysostaphin on sow endometritis (82.5%) was greater than that of the antibiotic group (72.17%) [3]. Phage lysin and chimeric lysin, as novel antibiotic alternatives, can rapidly lyse bacteria and not induce bacterial resistance to phage lysin [14,15,16]. At present, many bacterial diseases, including endometritis, osteomyelitis, and bovine mastitis, are induced by various bacterial species. However, to our knowledge, it is difficult for a phage lysin to efficiently lyse multiple genera of bacteria. For example, the S. aureus phage lysin LysGH15 has potent activity specifically targeting Staphylococcus spp. but has no discernible antibacterial efficacy against other bacterial strains [17,18,19]. Similarly, the chimeric lysin Csl2 possesses antibacterial activity against Streptococcus but not S. aureus [20]. To address these challenges, we envision that novel chimeric lysins can be exploited and applied to address the lack of host range of parental phage lysins. The chimeric lysin and parental phage lysin can be composed of a phage lysin cocktail against multiple bacterial infections. Furthermore, the broad-spectrum activity of antibiotics can be a suitable component. It has been widely proposed that the combination of phage lysin and antibiotics is superior to phage lysin and antibiotic monotherapy for treating bacterial infections [21,22,23] and can be applied for the treatment of bacterial infections in humans and animals [24, 25]. Additionally, our previous study revealed that the chimeric lysin ClyQ does not induce resistance in S. aureus and that the combination of ClyQ and mupirocin can delay the development of mupirocin resistance [26]. In conclusion, the combination of a phage lysin cocktail and antibiotics may be a solution for multibacterium-induced infections. However, the efficacy of a combination of antibiotics and phage lysin cocktails for the prevention of bacterium-induced endometritis remains to be elucidated.

In this study, a total of 526 bacteria, including Staphylococcus spp. (26.3%), Streptococcus spp. (12.3%), E. coli (28.9%), Enterococcus spp. (20.1%), Proteus spp. (9.5%), and Corynebacterium spp. (2.8%), were isolated from 383 vaginal swab samples from sows with endometritis, and the antibiotic resistance of Staphylococcus and Streptococcus was tested. Moreover, we constructed and purified a novel broad-host-range chimeric lysin, ClyL, which possessed excellent antibacterial activity against Staphylococcus app. Moreover, the prophage lysin Lys0859 was reported in our previous study and was shown to have exceptional activity against Streptococcus app [27]. ClyL and Lys0859 are composed of a phage lysin cocktail and exhibit synergistic effects against dual-species biofilms, Galleria mellonella larvae, and mouse mastitis models caused by coinfection with Staphylococcus and Streptococcus. Finally, the phage lysin cocktail was combined with cefquinome to form a combination therapy. We evaluated the effects of combination therapy on the activity of semen and the effects of semen on the bactericidal activity of combination therapy. Additionally, to prevent sow endometritis, we utilized semen as a vector to transport the combination of phage lysin cocktail and cefquinome by artificial insemination (AI).

Materials and methods

Collection of sow endometritis samples and microbiological analysis

Sow endometritis samples were collected as previously described [4, 28]. A total of 383 vaginal swab samples were collected from sows suffering from endometritis on 24 large-scale sow farms between April and August 2022 in Guigang city, Guangxi Province, Southern China. The sows that developed endometritis within five days following their first to sixth deliveries were selected for this study. These sows presented symptoms of fever and anorexia, and their vulvas were erythematous and oedematous. Additionally, purulent or mucopurulent uterine exudates are present in the vaginal canal of affected sows [1, 2]. To avoid contamination of the vaginal samples during sampling, special swab equipment was used to collect the samples. The structure of the swab equipment was the same as previously described [4]. All the samples were immediately placed on dry ice and transported to the laboratory. Afterwards, the samples were plated on tryptic soy agar (TSA) plates, which were incubated at 37 °C for 24–48 h. Single colonies of various shapes and sizes were selected and purified.

The strains were inoculated into tryptic soy broth (TSB) and cultured for 18–24 h at 37 °C, after which the bacterial DNA was extracted using a Bacterial DNA Extraction Kit (Tiangen Biotech, China) following the manufacturer’s instructions (Tiangen Biotech, China). The concentrations of extracted DNA were determined using a spectrophotometer (Thermo Scientific, USA). All the DNA samples were stored at −20 °C until further use. The bacteria were identified by the previously described primer sequences (Additional file 1) and sequenced by Guangzhou Ige Biotechnology Ltd. (Guangzhou, China), and the sequences were compared with the 16S rDNA sequence in the NCBI (National Center for Biotechnology Information) public database using the BLAST program (Basic Local Alignment Search Tool). The serotypes of Streptococcus suis were determined as previously described [29]. The related primers are listed in Additional file 2.

Antimicrobial susceptibility testing

The minimal inhibitory concentrations (MICs) of the isolated E. coli, Staphylococcus, and Streptococcus strains were determined according to the Clinical Laboratory and Standards Institute (CLSI) 2018 [30], and the breakpoints of antibiotics against E. coli, Staphylococcus, and Streptococcus are presented in Additional file 3. A total of fifteen antibiotics were selected, including ampicillin, amoxicillin, penicillin G, ceftiofur, cefquinome, gentamicin, tetracycline, doxycycline, florfenicol, chloramphenicol, amikacin, erythromycin, tilmicosin, lincomycin, and enrofloxacin. S. aureus ATCC 29213 and Streptococcus pneumoniae ATCC 49619 served as quality control strains. Finally, the MIC50 and MIC90 values were calculated as the MIC that inhibited 50% and 90% of the isolates, respectively.

Expression and purification of ClyL and Lys0859

The protocols for constructing and purifying Lys0859 were reported in our previous study [27]. The gene encoding the full-length S. aureus phage lysin LysGH15 (GenBank: ADG26756.1) was synthesized by Tsingke Biotechnology Co., Ltd. ClyL was constructed from the putative catalytic domain CHAP (from LysGH15, amino acids 1–165) with a cell wall-binding domain (from Lys0859, amino acids 143–239). The primers used for the construction of ClyL are listed in Additional file 4. The expression and purification of ClyL were carried out as previously described [26]. Briefly, the pET-28a (+) _ClyL vector was transformed into E. coli BL21 (DE3) cells, which were subsequently grown to OD600nm ~ 0.6 in LB containing 50 μg/mL kanamycin (37 °C, 200 g). The BL21 cells were subsequently cultured at 16 °C with 0.8 mM isopropyl-beta-d-thiogalactopyranoside (IPTG) for 16–18 h and then harvested by centrifugation at 6000 × g for 15 min. The cells were subsequently resuspended in binding buffer (250 mM NaCl, 20 mM Tris–HCl, pH 7.4), and the supernatants were collected after sonication at 4 °C. Lys0859 was purified by binding the 6× His-tagged fusion protein supernatant to a HisTrap FF column (GE Healthcare Bio-Sciences AB, USA) and eluted with elution buffer (250 mM NaCl, 20 mM Tris–HCl, 200 mM imidazole, pH 7.4) following the manufacturer’s instructions (AKTA pure 25 M, GE Healthcare Bio-Sciences AB, USA).

Lytic activity of ClyL and Lys0859 toward Streptococcus and Staphylococcus

The lytic activity of ClyL and Lys0859 against Streptococcus and Staphylococcus was examined through turbidity reduction and bactericidal assays as previously described [27]. First, 100 μL of log-phase bacteria in PBS (OD600nm = 0.6–0.8) was added to 100 μL of ClyL or Lys0859 in 96-well plates (NEST Biotechnology Co., Ltd., China), with PBS used as a negative control. The final concentrations of ClyL and Lys0859 were 50 μg/mL. After incubation at 37 °C for 30 min, the OD600nm values of the mixtures, including bacteria and lysin, were measured. On the other hand, log-phase bacteria were added to ClyL and Lys0859 at 37 °C for 1 h, and the final concentrations of ClyL and Lys0859 were 50 μg/mL. Thereafter, the bacterial count in the suspension treated with lysin was calculated via plate counting of tenfold serial dilutions. All the experiments were performed in triplicate.

Checkerboard assay

The checkerboard assay for the combination of ClyL and Lys0859 was performed in 96-well plates as previously described [31]. The concentration gradients of ClyL and Lys0859 were prepared in the horizontal and vertical directions. The log-phase S. aureus strains ATCC29213, S. agalactiae ATCC13813, and Staphylococcus 65 were diluted to 105 cfu/mL, and the bacteria were added to each well. The plates were subsequently incubated at 37 °C for 16–18 h. Finally, the MIC was recorded. Moreover, the MIC of ClyL and Lys0859 alone against the above bacteria were also tested. The values of the fractional inhibitory concentration index (FICI) were calculated according to previous methods [32].

Antibiofilm activity of ClyL and Lys0859

The antibiofilm ability of ClyL and Lys0859 was evaluated as previously described, with some changes [27]. Staphylococcus and Streptococcus strains were diluted to 1.0 × 105 cfu/mL. Two hundred microliters of bacteria were subsequently added to 96-well plates, which were subsequently incubated for 24 and 48 h at 37 °C. Two hundred microlitres of PBS, ClyL (100 μg/mL), Lys0859 (100 μg/mL), ClyL (100 μg/mL) + Lys0859 (100 μg/mL), or ClyL (50 μg/mL) + Lys0859 (50 μg/mL) was then added to the 96-well plates and incubated at 37 °C for 2 h. The biofilm biomass was evaluated using crystal violet [1.0% (weight/vol)] staining, and the viable bacteria were counted on the TSB agar plates. In addition, to determine the number of viable bacteria in the dual-species biofilms, S. aureus ATCC29213 and S. agalactiae ATCC13813 were cultured on Baird‒Parker agar (BP, HuanKai Microbial, Guangzhou, China) and Edwards Medium agar (modified, Oxoid, UK), respectively [33]. Scanning electron microscopy (SEM) images of the dual-species biofilms were obtained as previously described [26]. Staphylococcus and Streptococcus strains were diluted to 1.0 × 105 cfu/mL. S. aureus ATCC29213 and S. agalactiae ATCC13813 were mixed equivalently, and then, the mixtures were grown in media (TSB: PBS = 1:1) on 24-well plates with coverslips at 37 °C for 48 h. Thereafter, the 24-well plates were treated with PBS, ClyL (100 μg/mL), Lys0859 (100 μg/mL), and phage lysin cocktail ClyL (100 μg/mL) + Lys0859 (100 μg/mL) at 37 °C for 2 h. Finally, the images of the dual-species biofilms were observed by FESEM (2000× and 5000×) (NTC, Japan).

The effectiveness of phage lysin cocktail on G. mellonella larvae and a mouse mastitis model of coinfection with S. aureus ATCC29213 and S. agalactiae ATCC13813

The G. mellonella larvae were injected with bacteria and phage lysin as previously described [34]. Larvae of G. mellonella were obtained from Guiling Jiacheng Co., Ltd. (Guangxi, China). To identify the minimum lethal dose (MLD100), G. mellonella larvae were infected with different doses of S. aureus ATCC29213 or S. agalactiae ATCC13813 (5 × 106, 1 × 106, 5 × 105, or 1 × 105 cfu/bacteria/larvae). The larvae were inoculated with S. aureus ATCC29213 and S. agalactiae ATCC13813 at a concentration of 5 × 106 cfu/bacteria/larvae, followed by treatment with a combination of ClyL (1.5 μg/larvae) and Lys0859 (1.5 μg/larvae), ClyL (1.5 μg/larvae), Lys0859 (1.5 μg/larvae), or PBS. The survival rates of the larvae were observed at 72 h post infection. A mouse model of bovine mastitis was generated as previously described with some modifications [35]. Briefly, the L4 and R4 mammary glands were injected with 1 × 105 cfu/g of S. aureus ATCC29213 or S. agalactiae ATCC13813. The mice were subsequently divided into four groups (n = 3), and the mice were injected with a combination of 20 μg/g of Lys0859 or ClyL, 20 μg/g of Lys0859, 20 μg/g of ClyL, or 50 μL of PBS at 24 h postinfection. The L4 and R4 mammary gland tissues were ground, serially diluted, and spread on TSA plates at 48 h postinfection.

Assessment of the effects of the combination of the lysin cocktail and cefquinome on bacterial activity in semen

The lytic activity of ClyL, Lys0859, and cefquinome against bacteria was assessed in semen and TSB as previously described [36]. Briefly, Staphylococcus 65 was grown to the exponential growth phase. The bacterial suspension was subsequently centrifuged and washed twice with PBS. Finally, the bacteria were resuspended in TSB and commercial boar semen (Guangxi Yangxiang Co., Ltd.) and diluted to 106 cfu/mL. Commercial boar semen includes boar semen and Zenolong® extender (Beikang, Taizhou, China). As commercial extenders, the specific composition of Zenolong® is not known. The concentration of sperm in commercial boar semen is 2.5 × 107 spermatozoa/mL [37]. With respect to the phage lysin cocktail and cefquinome used in isolation, ClyL, Lys0859, and cefquinome were added to TSB. Next, the concentrations of ClyL and Lys0859 were adjusted to 50, 25, 12.5, and 0 μg/mL, and those of cefquinome were adjusted to 4, 2, 1 (2 μg/mL), and 0 × MIC. For the combination of the phage lysin cocktail and cefquinome, the phage lysin cocktail ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL), cefquinome monotherapy (20 × MIC, 80 μg/mL), and ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL) + cefquinome (20 × MIC) were added to TSB and semen. TSB and semen containing PBS and Staphylococcus 65 served as the control group. The mixtures were subsequently incubated at 37 °C for 24 h. Finally, the total number of bacterial colonies was counted at 0, 2, 4, 6, 8, 12, 16, 20, and 24 h. All the experiments were repeated three times.

Interactions among phage lysin, cefquinome, and boar semen

The phage lysin cocktail ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL) alone, cefquinome (20 × MIC, 80 μg/mL) alone, and ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL) + cefquinome (20 × MIC, 80 μg/mL) were added to commercial boar semen. Next, the semen was stored at 17 °C. The antibacterial activity of the phage lysin cocktail alone, cefquinome alone, and ClyL + Lys0859 + cefquinome groups was examined at 4, 7, and 11 days. Specifically, the Staphylococcus 65 strain (106 cfu/mL) was added to semen containing phage lysin cocktail, cefquinome, and ClyL + Lys0859 + cefquinome and incubated at 37 °C for 2 h. The viable bacteria were counted on TSB agar plates. Moreover, the total motility, progressive motility, velocity curved line (VCL), average path velocity (VAP), straight-line velocity (VSL), linearity (LIN), amplitude of lateral head displacement (ALH), wobble (WOB), and beat-cross frequency (BCF) of the sperm were observed by computer-aided sperm analysis (CASA) (IMV Technologies, France) as previously described [38, 39]. The acrosome integrity and MMP of the sperm were determined by the Viability and Acrosome Integrity Easykit 5 (IMV Technologies, France) and Mitochondrial Activity EasyKit 2 (IMV Technologies, France), respectively, as previously described [40].

Efficacy of cefquinome combined with phage lysin cocktail in sow endometritis

Approximately 20 days postpartum, the sows were acquired from a commercial swine farm in Guangxi Province, China, and subsequently allocated into four experimental groups (n = 7). The veterinarian verified that these sows did not suffer from endometritis and only exhibited normal inflammation after delivery. The veterinarian also confirmed that the sows met the criteria for AI. The four groups of sows were treated with commercial semen twice within 24 h, which was mixed with PBS, ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL), cefquinome (80 μg/mL) alone, or ClyL (25 μg/mL) + Lys0859 (12.5 μg/mL) + cefquinome (80 μg/mL) via AI. The clinical pregnancy rate, live birth rate, and recurrence endometritis rate were recorded at 4 months after AI.

Statistical analysis

Statistical analyses were performed using GraphPad Prism (version 9). Statistical significance was determined using an unpaired Student’s t test. P < 0.05 was considered statistically significant. All the data are expressed as the means ± standard deviations (SD) of three independent experiments.

Results

Identification of sow endometritis isolates

A total of 526 bacterial strains were isolated from 383 vaginal swab samples. The 526 strains included Staphylococcus spp. (26.3%, 141/536), Streptococcus spp. (12.3%, 66/536), E. coli (28.9%, 155/536), Enterococcus spp. (20.1%, 108/536), Proteus spp. (9.5%, 51/536), and Corynebacterium spp. (2.8%, 15/536) (Table 1). Moreover, all the Streptococcus strains were identified as S. suis (Table 1). The proportions of all strains are listed in Table 1. Similarly, a previous study reported that the bacteria most frequently isolated from mares with endometritis were E. coli (17.3%), Staphylococcus spp. (15.6%), and Streptococcus spp. (13.5%) [41]. In addition, 556 strains, including S. aureus (34%, n = 188), Streptococcus (32%, n = 181), and E. coli (19%, n = 104), were collected from 200 sows infected with endometritis [3]. Taken together, the results of this study inferred that E. coli, Staphylococcus, and Streptococcus (S. suis) play a pivotal role in endometritis.

Table 1 Prevalence of bacteria isolated from sow endometritis

Antibiotic susceptibility of sow endometritis isolates

Our initial step is to identify an antibiotic that is sensitive to bacteria and can effectively synergize with phage lysin to combat endometritis induced by multiple bacterial strains. The results of bacterial sensitivity to antimicrobial drugs can guide the utilization of antibiotics [41]. Therefore, the MICs of the fifteen antibiotics were tested for Staphylococcus, S. suis, and E. coli. On the basis of the existing criteria for antibiotics (Additional file 3), 63.1% (89/141), 85.5% (121/141), and 88.7% (125/141) of the Staphylococcus strains were insensitive to chloramphenicol, erythromycin, and lincomycin, respectively (Additional file 5). The findings regarding antibiotic resistance revealed that a significant proportion of S. suis strains, specifically 98.5% (65/66), 93.9% (62/66), and 100% (66/66), exhibited resistance to tetracycline, erythromycin, and lincomycin, respectively (Additional file 6). A total of 81.3% (126/155), 71.6% (111/155), 62.6% (97/155), 69.7% (108/155), and 71.0% (110/155) of the E. coli were resistant to ampicillin, tetracycline, doxycycline, florfenicol and chloramphenicol, respectively (Additional file 7).

Construction and expression of the chimeric lysin ClyL and effects of ions on the enzymatic activity of ClyL

On the basis of the results of antibiotic susceptibility, E. coli, Staphylococcus, and Streptococcus (S. suis) isolated from sow endometritis samples were sensitive to cefquinome. Therefore, it is imperative to exploit phage lysin, which can efficiently lyse both Staphylococcus and S. suis, for the successful implementation of the proposed combined therapy. Our previous study demonstrated that Lys0859 displayed excellent activity against 15 serotypes of S. suis [27]. Moreover, the S. aureus phage lysin LysGH15 has exceptional effects on Staphylococcus [17,18,19, 42]. Hence, to obtain an antistaphylococcal phage lysin, we constructed a new chimeric lysin, ClyL, which is composed of the catalytic domain CHAP from LysGH15 and a CBD of the prophage lysin Lys0859 (Figure 1A). The molecular weight of ClyL was approximately 34.0 kDa (Figure 1B). The stability of the novel chimeric lysin ClyL was tested in the presence of various metal ions. The results of the turbidity reduction assay indicated that varying concentrations of Ca2+ (1–50 mM), Mg2+ (1–10 mM), and Zn2+ (1–50 mM) have the potential to enhance the lytic activity of ClyL, as depicted in Figure 1C-E. Additionally, a heightened bactericidal effect of ClyL was noted in PBS buffer with 5 mM concentrations of Ca2+, Mg2+, and Zn2+ (Figure 1F). Similarly, Ca2+ promotes the activity of the chimeric lysin ClyC in a dose-dependent manner (1–1000 μM) [43]. Research has shown that the lytic activity of the LysGH15CHAP domain is specifically dependent on calcium [18]. The findings of our study indicated that Mg2+, Zn2+, and Ca2+ were capable of enhancing the bactericidal efficacy of ClyL (Figure 1C–F). We previously reported that Lys0859 exhibited increased activity against S. suis in PBS buffer containing Mg2+ and Ca2+ but had little lytic activity in PBS buffer containing Zn2+ [27].

Figure 1
figure 1

Schematic representation and SDS analysis of ClyL. A The novel chimeric lysin ClyL, which is composed of a cysteine- and histidine-dependent amidohydrolase/peptidase (CHAP) catalytic domain (1–165 aa) from the phage lysin LysGH15 and a cell wall-binding domain (CBD) (143–239 aa) from the prophage lysin Lys0859. B SDS‒PAGE of the recombinant chimeric lysin ClyL. M: marker; 1: purified ClyL (34.0 kDa). CF Effects of metal ions on the antibacterial activity of Lys0859. Different concentrations of Ca2+ (C), Mg2+ (D), and Zn2+ (E) were added to ClyL (50 μg/mL), and then the mixtures were mixed with S. aureus ATCC 29213 at 37 °C for 30 min. E PBS and 5 mM Ca2+, Mg2+, and Zn2+ were added to Lys0859 (50 μg/mL), and the mixtures were mixed with S. aureus ATCC 29213 at 37 °C for 60 min. Significant differences between the PBS groups and the ClyL groups were determined by Student’s t test (*** P < 0.001). Significant differences between the ClyL groups and the ClyL + 5 mM Ca2+, ClyL + 5 mM Mg2+, and ClyL + 5 mM Zn2+ groups were determined by Student’s t test (# P < 0.05, ## P < 0.05). The data are expressed as the mean ± standard deviation (SD).

Staphylococcus and Streptococcus strains isolated from sow endometritis and bovine mastitis samples are sensitive to ClyL and Lys0859 in vitro

First, S. suis and Staphylococcus strains isolated from sow endometritis were utilized to assess the lytic activity of ClyL and Lys0859. As depicted in Figure 2A and C, the turbidity of Staphylococcus (85.2%) and S. suis (95.4%) could be decreased by 20% or more by ClyL. Similarly, the turbidity of Staphylococcus (48.9%) and S. suis (100%) could be decreased by greater than 20% by Lys0859 (Figure 2B and D). Details of turbidity are shown in Additional files 8 and 9. In addition, bovine mastitis is induced by multiple bacterial species, and staphylococcal and streptococcal strains are the main bacterial pathogens [44]. Thus, we utilized staphylococcal and streptococcal strains from bovine mastitis to determine the bactericidal activity of ClyL and Lys0859. As shown in Figure 2E, ClyL showed exceptional antistaphylococcal activity compared with Lys0859. Moreover, compared with ClyL, Lys0859 exhibited significant antibacterial activity against Streptococcus (Figure 2E). Similarly, according to the bacterial count test, the activities of ClyL and Lys0859 were most pronounced for the Staphylococcus and Streptococcus strains isolated from sow endometritis and bovine mastitis, respectively (Figure 2F and G). To determine the synergistic effect of ClyL and Lys0859, we tested the FICI values of a combination of ClyL and the parental phage lysin Lys0859. The fractional inhibitory concentration index (FICI) values for S. aureus ATCC29213, S. agalactiae ATCC13813, and Staphylococcus 65 were determined to be 0.5, 0.266, and 0.266, respectively (Additional file 10). For the coinfection of S. aureus ATCC29213 and S. agalactiae ATCC13813, the FICI value was 0.281 (Additional file 10). On the basis of the established criteria for synergistic effects outlined previously [26, 32], ClyL and Lys0859 exhibit synergistic activity against strains of Staphylococcus and Streptococcus, as well as coinfections involving these bacterial strains.

Figure 2
figure 2

The phages lysin ClyL and Lys0859 could effectively lyse bacteria isolated from sows with endometritis and bovine mastitis. AD The antibacterial activity of Lys0859 (50 μg/mL) and ClyL (50 μg/mL) against clinical bacteria isolated from sows subjected to endometritis at 37 °C for 1 h. The values of turbidity decrease were classified into three parts: a decrease in turbidity > 50%, a 20% ≤ decrease in turbidity ≤ 50%, and a decrease in turbidity < 20%. The proportions of the three parts are presented in a circular figure. The proportions of the decrease in ClyL turbidity against Staphylococcus and Streptococcus are listed in A and C, respectively. The proportions of Lys0859 turbidity decreases against Staphylococcus and Streptococcus are listed in B and D, respectively. E The antibacterial activity of Lys0859 (50 μg/mL) and ClyL (50 μg/mL) against clinical bacteria isolated from bovine mastitis. F, G The bacterial count test was used to evaluate the bactericidal activity of Lys0859 (50 μg/mL) and ClyL (50 μg/mL) against Staphylococcus and Streptococcus strains isolated from sows with endometritis (F) and bovine mastitis (G). Significant differences between the Lys0859 groups and ClyL groups were determined by Student’s t test (** P < 0.01, and *** P < 0.001). The data represent the mean ± SD of three independent experiments.

The combination of ClyL and Lys0859 has a synergistic effect on coinfection with Staphylococcus and Streptococcus in vitro and in vivo

At present, we have demonstrated that ClyL and Lys0859 display the highest levels of activity against Staphylococcus and Streptococcus strains, resulting in a synergistic bactericidal effect. Numerous studies indicate that bovine mastitis is attributed to multiple bacterial infections [45]. In bovine mastitis, S. agalactiae and S. aureus play major roles in the formation of biofilms and implantation in the mammary gland [46, 47]. The models of dual-species biofilms, Galleria mellonella larvae, and mastitis in mice were used to evaluate the synergistic effect against coinfection with Staphylococcus and Streptococcus in vitro and in vivo. First, we determined that ClyL and Lys0859 could clear S. aureus ATCC29213 and S. agalactiae ATCC13813 biofilms (Additional file 11). Thereafter, we constructed a dual-species biofilm model. The biomass and bacterial count of the dual-species biofilms were greatest when 1 × 105 cfu/mL S. aureus ATCC29213 or S. agalactiae ATCC13813 was cultured in 50% TSB (TSB: PBS = 1:1) for 24 h (Figure 3A–C). The dual-species biofilms of S. aureus ATCC29213 and S. agalactiae ATCC13813 were subsequently cultured at 37 °C for 24 h and 48 h. Our results indicated that a combination of ClyL (100 μg/mL) and Lys0859 (100 μg/mL) significantly decreased the biomass and bacterial number of dual-species biofilms compared with those in the PBS groups (P < 0.01) (Figure 3D and E). The combination of ClyL (100 μg/mL) and Lys0859 (100 μg/mL) or ClyL (50 μg/mL) and Lys0859 (50 μg/mL) was superior to ClyL (100 μg/mL) or Lys0859 (100 μg/mL) alone for clearing biofilms (Figure 3D and E). Moreover, there was no significant difference in antibiofilm ability between ClyL (100 μg/mL) + Lys0859 (100 μg/mL) and ClyL (50 μg/mL) + Lys0859 (50 μg/mL) (Figure 3D and E). The FESEM images of the dual-species biofilms were also consistent with the above results (Figure 3F). To explore the therapeutic effects of a combination of ClyL and Lys0859 in vivo, we developed a coinfection model using G. mellonella larvae infected with S. aureus ATCC29213 and S. agalactiae ATCC13813, as well as a mouse mastitis model. The survival rates of larvae treated with a combination of ClyL (1.5 μg/larvae) and Lys0859 (1.5 μg/larvae), ClyL (1.5 μg/larvae), Lys0859 (1.5 μg/larvae), and PBS were 80%, 35%, 30%, and 30%, respectively (Figure 4A). Previous work has confirmed that intramammary injection of Lys0859 (20 μg/gland) reduces the bacterial burden in mammary tissue by 3.74 logs [27]. In this study, the S. aureus and S. agalactiae numbers in mammary gland tissue were significantly lower in the ClyL (40 μg/mouse) and Lys0859 (40 μg/mouse) groups than in the PBS groups (P < 0.05) (Figure 4B). The combination of ClyL and Lys0859 was superior to the combination of ClyL or Lys0859 alone in eradicating bacteria from S. aureus ATCC29213-S. agalactiae ATCC13813-coinfected mammary gland tissue (Figure 4B). Together, the combined use of ClyL and Lys0859 may represent a viable approach for addressing coinfections caused by Staphylococcus and Streptococcus.

Figure 3
figure 3

Chimeric lysin ClyL and parental phage lysin Lys0859 were used to treat 24-h- and 48-h-old biofilms formed by S. aureus ATCC29213 and S. agalactiae ATCC13813 coinfection. AC The optimal conditions for dual-species biofilm formation. A The biomass of dual-species (S. aureus ATCC29213 and S. agalactiae ATCC13813) biofilms formed at 37 °C for 24 h. The viable cell counts of dual-species biofilms of S. aureus ATCC29213 (B) and S. agalactiae ATCC13813 (C) formed at 37 °C for 24 h. D The biomass of dual-species biofilms treated with ClyL, Lys0859, or ClyL + Lys0859 at 37 °C for 2 h. E Viable cell counts of dual-species biofilms treated with ClyL, Lys0859, and ClyL + Lys0859 at 37 °C for 2 h. Significant differences between the PBS groups and the ClyL, Lys0859, and ClyL + Lys0859 groups were determined via Student’s t test (ns P > 0.05, * P < 0.05 ** p < 0.01, and *** P < 0.001). Significant differences between the ClyL + Lys0859 groups and the ClyL and Lys0859 groups were determined via Student’s t test (ns P > 0.05, # P < 0.05, ## P < 0.01, and ### P < 0.001). The error bars represent the standard deviations (SD). F FESEM images of dual-species biofilms (48 h) formed by S. aureus ATCC29213 and S. agalactiae ATCC13813. The dual-species biofilms were treated with PBS, ClyL (100 μg/mL), Lys0859 (100 μg/mL), or the phage lysin cocktail ClyL (100 μg/mL) + Lys0859 (100 μg/mL).

Figure 4
figure 4

Effect of phage lysin cocktail treatment on S. aureus- and S. agalactiae-induced coinfections. A The therapeutic effect of the phage lysin cocktail on G. mellonella larvae coinfected with S. aureus ATCC29213 and S. agalactiae ATCC13813. The larvae were infected with a mixture of S. aureus ATCC29213 and S. agalactiae ATCC13813 and then treated with a phage lysin cocktail, chimeric lysin ClyL, or phage lysin Lys0859 at 3 h post infection. B The therapeutic effect of the phage lysin cocktail on S. aureus ATCC29213- and S. agalactiae ATCC13813-induced mastitis in mice. The mice were treated with phage lysin cocktail, chimeric lysin ClyL, or phage lysin Lys0859 at 24 h post infection. Significant differences between the PBS groups and the ClyL + Lys0859 groups were determined by Student’s t test (* P < 0.05 and ** P < 0.01). Significant differences between the ClyL + Lys0859 groups and the ClyL and Lys0859 groups were determined via Student’s t test (ns P > 0.05, ## P < 0.01, and ### P < 0.001). The error bars represent the standard deviations (SD).

The combination of phage lysin cocktail and cefquinome has a synergistic bactericidal effect in vitro

Notably, antibiotics and lysin have been theorized to destroy bacteria in distinct ways [48, 49], and a combination of antibiotics and lysin is beneficial for reducing the occurrence of bacterial resistance during treatment [26]. Unexpectedly, in this study, a combination of ClyL and Lys0859 swiftly lowered the bacterial count within 0–8 h but did not affect bacterial growth within 8–24 h (Figure 5A and B). In contrast, cefquinome consistently decreased the visible number of Staphylococcus 65 colonies within 24 h, although more slowly than ClyL and Lys0859 did (Figure 5A and B). Our previous study corroborated that the combination of the chimeric lysin ClyQ and mupirocin had a synergistic bactericidal effect on S. aureus strains [26]. Therefore, the ClyL + Lys0859, cefquinome alone, and cefquinome + ClyL + Lys0859 groups were designed to explore the synergistic effect of the phage lysin cocktail and cefquinome on bacterial endometritis. As depicted in Figure 5, the combination of phage lysin cocktail and cefquinome in TSB and semen was superior to ClyL + Lys0859 and cefquinome monotherapy in eliminating and inhibiting bacterial proliferation. ClyL and Lys0859 were utilized in the development of a phage lysin cocktail, which was subsequently combined with cefquinome to create a combined therapy for preventing sow endometritis after AI.

Figure 5
figure 5

The antibacterial activity of the combination of phage lysin cocktail and cefquinome in culture medium and semen. The antibacterial activities of the phage lysin cocktail alone, cefquinome alone, and the combination of cefquinome and phage lysin cocktail were analysed in culture medium (A) and semen (B). PBS was used as a negative control. Staphylococcus 65 was added to the culture medium and semen. The bacterial strains of Staphylococcus 65 in the culture medium and semen were incubated at 37 °C for 24 h and tested by plating on TSB agar at different times. Significant differences between the ClyL + Lys0859 groups and the Cefquinome + ClyL + Lys0859 groups were determined via Student’s t test (ns P > 0.05, * P < 0.05, and ** P < 0.01). Significant differences between the cefquinome groups and the cefquinome + ClyL + Lys0859 groups were determined by Student’s t test (# P < 0.05, ## P < 0.01, and ### P < 0.001). All the experiments were repeated three times. The error bars represent the standard deviation (SD).

Boar semen containing phage lysin and antibiotics is a viable strategy for preventing bacterial endometritis after AI

Because bacteria often infect boar semen during collection, processing, and storage and boar semen infected with bacteria can result in sow endometritis AI [50, 51], boar semen containing a phage lysin cocktail and cefquinome was used to control bacteria from semen and prevent sow endometritis via AI in this study. Next, the effects of phage lysin and cefquinome on the activity of semen were analysed, and the results indicated that, compared with those of the control group, the activities (including motility, acrosomal integrity, progressive motility, and mitochondrial membrane potential (MMP)) of semen were not significantly affected by cefquinome, the lysins ClyL and Lys0859 (Figure 6A–D, Additional file 12). Furthermore, compared with PBS, semen did not significantly influence the antibacterial activity of cefquinome, ClyL, or Lys0859 (Figure 6E). Compared with the control conditions, the combination of cefquinome, ClyL, and Lys0859 significantly reduced the number of bacteria in PBS and boar semen (Figure 6E). Importantly, the combination of cefquinome, ClyL, and Lys0859 significantly reduced the bacterial number in boar semen compared with that in the ClyL + Lys0859 and cefquinome alone groups (Figure 6E).

Figure 6
figure 6

Effects of ClyL, Lys0859, cefquinome and boar sperm. AD Effects of ClyL, Lys0859, and cefquinome on the activity of boar sperm. The concentrations of ClyL and Lys0859 were 25 and 12.5 μg/mL, respectively. The cefquinome concentration was 20 × MIC (80 μg/mL). PBS, cefquinome, ClyL + Lys0859, and cefquinome + ClyL + Lys0859 were added to boar semen. The semen was subsequently stored at 17 °C, and the total motility (A), acrosomal integrity (B), progressive motility (C), and mitochondrial membrane potential (MMP) (D) of the sperm were monitored on the 4th, 8th, and 12th days of storage. (E) boar semen did not significantly affect the antibacterial activity of ClyL, Lys0859, or cefquinome. The PBS, cefquinome, ClyL + Lys0859, and cefquinome + ClyL + Lys0859 were mixed with PBS and boar semen. The mixtures were then stored at 17 °C, and the lytic activities of PBS, cefquinome, ClyL + Lys0859, and cefquinome + ClyL + Lys0859 in PBS and semen against Staphylococcus 65 were monitored on the 4th, 7th, and 11th days of storage. The ClyL + Lys0859, cefquinome, cefquinome + ClyL + Lys0859 and PBS groups were compared via Student’s t test (**** P < 0.0001). The ClyL + Lys0859, cefquinome and cefquinome + ClyL + Lys0859 groups were compared via Student’s t test (ns P > 0.05, # P < 0.05, ### P < 0.001). The error bars represent the standard deviation (SD).

Combination therapy with phage lysin cocktail and cefquinome reduces the incidence rate of endometritis

Our results revealed that the incidence rate of sow endometritis was 0% (0/7) when the combination of a lysin cocktail and cefquinome was used compared with 50% (3/6) when PBS or cefquinome alone was administered (Table 2). In addition, the incidence rate of sow endometritis was 57.14% (4/7) when the phage lysin cocktail alone was administered. The clinical pregnancy and live birth rates of the PBS-treated group were 85.71% (6/7), and those of the combination-treated group were 100% (7/7) (Table 2). The average cure rates of lysostaphin and oxytetracycline for sow endometritis are 82.5% and 72.17%, respectively [3]. The results revealed that the combination of a phage lysin cocktail and antibiotics is a promising strategy for preventing endometritis in sows. Moreover, one-third of infertile women with RIF suffer from CE caused by common bacteria (Streptococcus, Staphylococcus, E. coli, and Enterococcus faecalis), and the success rate of embryo transfer in infertile couples can be influenced by RIF [52,53,54,55]. Thus, our findings demonstrated that combined therapy with a phage lysin cocktail and antibiotics can be applied for the treatment of women endometritis in future clinical practice.

Table 2 Reproductive outcomes of sows with endometritis in the PBS, ClyL + Lys0859, cefquinome, and cefquinome + ClyL + Lys0859 groups

Discussion

The results of this study suggest that E. coli, Staphylococcus, and Streptococcus (S. suis) play pivotal roles in endometritis. Moreover, antibiotics exhibit wide-ranging antibacterial properties, and phage lysin has been shown to rapidly lyse Staphylococcus and S. suis, with the exception of E. coli [15, 24]. Combined therapy with phage lysin and antibiotics delays the development of antibiotic resistance [26]. Hence, we developed a novel therapeutic approach that integrates and leverages the benefits of antibiotics and phage lysins for the treatment of bacterium-induced endometritis. Moreover, most cases of endometritis are polymicrobial and involve aerobic and anaerobic bacteria. Research has shown that obligate anaerobes, including Peptoniphilus, Bacteroides, and Clostridium, are associated with dairy cattle endometritis [56]. The isolation procedure did not consider anaerobic cultivation conditions, which resulted in a lack of information on the obligate anaerobes. In this study, MIC50 values were also used to choose the ideal antibiotic owing to the paucity of existing criteria for antibiotics. On the basis of the results of antibiotic resistance, the MIC50 values of cefquinome for Staphylococcus, S. suis, and E. coli were 1, 0.06, and 0.12 μg/mL, respectively (Additional files 5, 6, 7). In addition, the MIC90 value of cefquinome for both Staphylococcus and S. suis was 2 μg/mL, which was the lowest among all antibiotics (Additional files 5 and 6). Hence, cefquinome was the most suitable antibiotic against bacteria causing sow endometritis in this study. However, cefquinome, classified as a fourth-generation cephalosporin, is considered a critically important antibiotic for human health according to the World Health Organization (WHO) [57]. Enrofloxacin, an FDA- and China-approved fluoroquinolone, has been used broadly for the treatment of swine disease caused by Gram-positive and Gram-negative bacteria [58, 59]. In this study, the MIC50 values of enrofloxacin for Staphylococcus, S. suis, and E. coli were 1, 0.25, and 1 μg/mL, respectively (Additional files 5, 6, 7). Therefore, enrofloxacin may be a more suitable antibiotic for treating sow endometritis than cefquinome in the future.

Thus, it is imperative to exploit phage lysin, which can efficiently lyse both Staphylococcus and S. suis, for the successful implementation of the proposed combined therapy. Our results implied that ClyL and Lys0859 could effectively lyse and exhibited synergistic activity against Staphylococcus and Streptococcus strains in sows with endometritis and bovine mastitis. An earlier study demonstrated that phage lysin and cefquinome can treat bacterial infections in swine [60, 61] but not sow endometritis. This study represents the first report of the use of a combination of phage lysin and antibiotic treatment to address sow endometritis, with no observed significant impact on semen quality. However, combination therapy does not eliminate the use of antibiotics. E. coli, Staphylococcus, and S. suis play vital roles in endometritis in this study. ClyL and Lys0859 can lyse Staphylococcus and S. suis, with the exception of E. coli. There have been reports on the use of the E. coli phages lysin PlyE146 and Lysep3, which display antimicrobial activity towards E. coli [62, 63]. Thus, we can develop a phage lysin that targets E. coli to eliminate the use of cefquinome. Taken together, the results revealed that semen containing phage lysin and antibiotics was a viable strategy for preventing bacterial endometritis.

Research has shown that antibiotic therapy can be used for the treatment of endometritis in both animals and women [10, 64]. Although antibiotic therapy resulted in chronic endometritis resolution in 82.3% of patients, 17.6% of patients experienced persistent endometritis [65]. These challenges highlight the importance of diverse antimicrobial strategies. At present, phage lysin has been successfully applied for the treatment of various bacterial infections [31, 66]. The incidence rate of sow endometritis was 57.14% (4/7) when the phage lysin cocktail alone was administered. The clinical pregnancy and live birth rates of the PBS-treated group were 85.71% (6/7), and those of the combination-treated group were 100% (7/7). In accordance with the findings presented above, to address the limitations of antibiotic monotherapy, we developed a potential strategy in which semen was employed as a carrier to deliver a combination of phage lysin cocktail and cefquinome through AI.

In brief, 526 strains consisting of Staphylococcus spp. (26.3%, 141/536), Streptococcus spp. (12.3%, 66/536), E. coli (28.9%, 155/536), Enterococcus spp. (20.1%, 108/536), Proteus spp. (9.5%, 51/536), and Corynebacterium spp. (2.8%, 15/536) were isolated from sow endometritis samples. According to the findings of antibiotic susceptibility testing, Staphylococcus, Streptococcus, and E. coli strains isolated from sow endometritis samples demonstrated sensitivity to cefquinome. A novel antimicrobial agent, the chimeric lysin ClyL, was subsequently developed and demonstrated remarkable bactericidal efficacy against Staphylococcus strains obtained from cases of sow endometritis and bovine mastitis. Moreover, the phage lysin Lys0859 had significant effects on Streptococcus strains isolated from sows with endometritis and bovine mastitis. ClyL and Lys0859 were used to design a phage lysin cocktail that exhibited synergistic effects against coinfection with Staphylococcus and Streptococcus in vitro and in vivo. Thus, we conceived a novel therapy that combines cefquinome and phage lysin cocktails containing ClyL and Lys0859 against bacterium-induced sow endometritis. The combination of cefquinome and the phage lysin cocktail did not affect sperm activity but had a synergistic bactericidal effect on semen. Additionally, semen did not significantly influence the bactericidal activity of the phage lysin cocktail combined with cefquinome. Furthermore, this novel approach reduces the incidence rate of sow endometritis by using semen as a vector to deliver a phage lysin cocktail and cefquinome via AI. In conclusion, we speculate that this novel strategy could be applied to prevent endometritis after AI and coinfections with Staphylococcus and Streptococcus.

Availability of data and materials

The datasets used and analysed during the current study are available from the corresponding author upon reasonable request.

References

  1. Zhang L, Wang L, Dai Y, Tao T, Wang J, Wu Y, Zeng X, Zhang J (2021) Effect of sow intestinal flora on the formation of endometritis. Front Vet Sci 8:663956

    Article  PubMed  PubMed Central  Google Scholar 

  2. Wang Y, Guo H, Bai Y, Li T, Xu R, Sun T, Lu J, Song Q (2020) Isolation and characteristics of multi-drug resistant Streptococcus porcinus from the vaginal secretions of sow with endometritis. BMC Vet Res 16:146

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Ye G, Huang J, Li G, Zhang J, Sun Y, Zeng D, Bao W, Zhong J, Huang Q (2021) Clinical efficacy of intravaginal recombinant lysostaphin administration on endometritis in sows. Vet Med Sci 7:746–754

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Wang J, Li C, Nesengani LT, Gong Y, Zhang S, Lu W (2017) Characterization of vaginal microbiota of endometritis and healthy sows using high-throughput pyrosequencing of 16S rRNA gene. Microb Pathog 111:325–330

    Article  PubMed  CAS  Google Scholar 

  5. Lager KM, Halbur PG (1996) Gross and microscopic lesions in porcine fetuses infected with porcine reproductive and respiratory syndrome virus. J Vet Diagn Invest 8:275–282

    Article  PubMed  CAS  Google Scholar 

  6. Jana B, Kucharski J, Dzienis A, Deptuła K (2007) Changes in prostaglandin production and ovarian function in gilts during endometritis induced by Escherichia coli infection. Anim Reprod Sci 97:137–150

    Article  PubMed  CAS  Google Scholar 

  7. Singh N, Sethi A (2022) Endometritis—diagnosis, Treatment and its impact on fertility—a scoping review. JBRA Assist Reprod 26:538–546

    PubMed  PubMed Central  Google Scholar 

  8. Barnes M, Kasimanickam R, Kasimanickam V (2023) Effect of subclinical endometritis and flunixin meglumine administration on pregnancy in embryo recipient beef cows. Theriogenology 201:76–82

    Article  PubMed  CAS  Google Scholar 

  9. Chagnon M, D’Allaire S, Drolet R (1991) A prospective study of sow mortality in breeding herds. Can J Vet Res 55:180–184

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Cicinelli E, Matteo M, Tinelli R, Lepera A, Alfonso R, Indraccolo U, Marrocchella S, Greco P, Resta L (2015) Prevalence of chronic endometritis in repeated unexplained implantation failure and the IVF success rate after antibiotic therapy. Hum Reprod 30:323–330

    Article  PubMed  Google Scholar 

  11. Pycock JF, Newcombe JR (1996) Assessment of the effect of three treatments to remove intrauterine fluid on pregnancy rate in the mare. Vet Rec 138:320–323

    Article  PubMed  CAS  Google Scholar 

  12. Salmanov AG, Vitiuk AD, Zhelezov D, Bilokon O, Kornatska AG, Dyndar OA, Trokhymovych OV, Bozhko N, Raksha II, Nykoniuk TR, Gorbunova OV, Kokhanov IV, Kushnirenko S, Iarotska I, Golianovsky OV, Holovanova IA, Abbasova ER (2020) Prevalence of postpartum endometritis and antimicrobial resistance of responsible pathogens in ukraine: results a multicenter study (2015–2017). Wiad Lek 73:1177–1183

    Article  PubMed  Google Scholar 

  13. Mahmoud SF, Fayez M, Swelum AA, Alswat AS, Alkafafy M, Alzahrani OM, Alsunaini SJ, Almuslem A, Al Amer AS, Yusuf S (2022) Genetic diversity, biofilm formation, and antibiotic resistance of Pseudomonas aeruginosa isolated from cow, camel, and mare with clinical endometritis. Vet Sci 9:239

    Article  PubMed  PubMed Central  Google Scholar 

  14. Daniel A, Euler C, Collin M, Chahales P, Gorelick KJ, Fischetti VA (2010) Synergism between a novel chimeric lysin and oxacillin protects against infection by methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 54:1603–1612

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Gilmer DB, Schmitz JE, Euler CW, Fischetti VA (2013) Novel bacteriophage lysin with broad lytic activity protects against mixed infection by Streptococcus pyogenes and methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 57:2743–2750

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Yang H, Gong Y, Zhang H, Etobayeva I, Miernikiewicz P, Luo D, Li X, Zhang X, Dąbrowska K, Nelson DC, He J, Wei H (2019) ClyJ is a novel pneumococcal chimeric lysin with a cysteine- and histidine-dependent amidohydrolase/peptidase catalytic domain. Antimicrob Agents Chemother 63:e02043-18

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Gu J, Xu W, Lei L, Huang J, Feng X, Sun C, Du C, Zuo J, Li Y, Du T, Li L, Han W (2011) LysGH15, a novel bacteriophage lysin, protects a murine bacteremia model efficiently against lethal methicillin-resistant Staphylococcus aureus infection. J Clin Microbiol 49:111–117

    Article  PubMed  CAS  Google Scholar 

  18. Gu J, Feng Y, Feng X, Sun C, Lei L, Ding W, Niu F, Jiao L, Yang M, Li Y, Liu X, Song J, Cui Z, Han D, Du C, Yang Y, Ouyang S, Liu ZJ, Han W (2014) Structural and biochemical characterization reveals LysGH15 as an unprecedented “EF-hand-like” calcium-binding phage lysin. PLoS Pathog 10:e1004109

    Article  PubMed  PubMed Central  Google Scholar 

  19. Zhang Y, Cheng M, Zhang H, Dai J, Guo Z, Li X, Ji Y, Cai R, Xi H, Wang X, Xue Y, Sun C, Feng X, Lei L, Han W, Gu J (2018) Antibacterial effects of phage lysin LysGH15 on planktonic cells and biofilms of diverse staphylococci. Appl Environ Microbiol 84:e00886-18

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Vázquez R, Domenech M, Iglesias-Bexiga M, Menéndez M, García P (2017) Csl2, a novel chimeric bacteriophage lysin to fight infections caused by Streptococcus suis, an emerging zoonotic pathogen. Sci Rep 7:16506

    Article  PubMed  PubMed Central  Google Scholar 

  21. Schuch R, Lee HM, Schneider BC, Sauve KL, Law C, Khan BK, Rotolo JA, Horiuchi Y, Couto DE, Raz A, Fischetti VA, Huang DB, Nowinski RC, Wittekind M (2014) Combination therapy with lysin CF-301 and antibiotic is superior to antibiotic alone for treating methicillin-resistant Staphylococcus aureus-induced murine bacteremia. J Infect Dis 209:1469–1478

    Article  PubMed  CAS  Google Scholar 

  22. Vouillamoz J, Entenza JM, Giddey M, Fischetti VA, Moreillon P, Resch G (2013) Bactericidal synergism between daptomycin and the phage lysin Cpl-1 in a mouse model of pneumococcal bacteraemia. Int J Antimicrob Agents 42:416–421

    Article  PubMed  CAS  Google Scholar 

  23. Torres-Barceló C (2018) The disparate effects of bacteriophages on antibiotic-resistant bacteria. Emerg Microbes Infect 7:168

    Article  PubMed  PubMed Central  Google Scholar 

  24. Fowler VG Jr, Das AF, Lipka-Diamond J, Schuch R, Pomerantz R, Jáuregui-Peredo L, Bressler A, Evans D, Moran GJ, Rupp ME, Wise R, Corey GR, Zervos M, Douglas PS, Cassino C (2020) Exebacase for patients with Staphylococcus aureus bloodstream infection and endocarditis. J Clin Invest 130:3750–3760

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Asempa TE, DeRosa NA, Cassino C, Lehoux D, Schuch R, Nicolau DP (2021) Efficacy assessment of lysin CF-296 in addition to daptomycin or vancomycin against Staphylococcus aureus in the murine thigh infection model. J Antimicrob Chemother 76:2622–2628

    Article  PubMed  CAS  Google Scholar 

  26. Duan XC, Li XX, Li XM, Wang S, Zhang FQ, Qian P (2023) Exploiting broad-spectrum chimeric lysin to cooperate with mupirocin against Staphylococcus aureus-induced skin infections and delay the development of mupirocin resistance. Microbiol Spectr 11:e0505022

    Article  PubMed  Google Scholar 

  27. Li XX, Zhang FQ, Wang S, Duan XC, Hu DY, Gao DY, Tao P, Li XM, Qian P (2023) Streptococcus suis prophage lysin as a new strategy for combating streptococci-induced mastitis and Streptococcus suis infection. J Antimicrob Chemother 78:747–756

    Article  PubMed  CAS  Google Scholar 

  28. Liang H, Cai R, Li C, Glendon OHM, Chengcheng H, Yan H (2022) High-throughput sequencing of 16S rRNA gene analysis reveals novel taxonomic diversity among vaginal microbiota in healthy and affected sows with endometritis. Res Vet Sci 143:33–40

    Article  PubMed  CAS  Google Scholar 

  29. Okura M, Lachance C, Osaki M, Sekizaki T, Maruyama F, Nozawa T, Nakagawa I, Hamada S, Rossignol C, Gottschalk M, Takamatsu D (2014) Development of a two-step multiplex PCR assay for typing of capsular polysaccharide synthesis gene clusters of Streptococcus suis. J Clin Microbiol 52:1714–1719

    Article  PubMed  PubMed Central  Google Scholar 

  30. CLSI (2020) Performance standards for antimicrobial susceptibility testing CLSI document. Ma100 Clinical Laboratory Standards Institute (CLSI), Wayne

    Google Scholar 

  31. Kim NH, Park WB, Cho JE, Choi YJ, Choi SJ, Jun SY, Kang CK, Song KH, Choe PG, Bang JH, Kim ES, Park SW, Kim NJ, Oh MD, Kim HB (2018) Effects of phage endolysin SAL200 combined with antibiotics on Staphylococcus aureus infection. Antimicrob Agents Chemother 62:e00731-18

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Shao Z, Wulandari E, Lin RCY, Xu J, Liang K, Wong EHH (2022) Two plus One: combination therapy tri-systems involving two membrane-disrupting antimicrobial macromolecules and antibiotics. ACS Infect Dis 8:1480–1490

    Article  PubMed  CAS  Google Scholar 

  33. Skarbye AP, Thomsen PT, Krogh MA, Svennesen L, Østergaard S (2020) Effect of automatic cluster flushing on the concentration of Staphylococcus aureus in teat cup liners. J Dairy Sci 103:5431–5439

    Article  PubMed  CAS  Google Scholar 

  34. Li X, Chen Y, Wang S, Duan X, Zhang F, Guo A, Tao P, Chen H, Li X, Qian P (2022) Exploring the benefits of metal ions in phage cocktail for the treatment of methicillin-resistant Staphylococcus aureus (MRSA) infection. Infect Drug Resist 15:2689–2702

    Article  PubMed  PubMed Central  Google Scholar 

  35. Brouillette E, Grondin G, Lefebvre C, Talbot BG, Malouin F (2004) Mouse mastitis model of infection for antimicrobial compound efficacy studies against intracellular and extracellular forms of Staphylococcus aureus. Vet Microbiol 101:253–262

    Article  PubMed  CAS  Google Scholar 

  36. Chang Y, Li Q, Zhang S, Zhang Q, Liu Y, Qi Q, Lu X (2023) Identification and molecular modification of Staphylococcus aureus bacteriophage lysin LysDZ25. ACS Infect Dis 9:497–506

    Article  PubMed  Google Scholar 

  37. Xiong Z, Hong Z, Li X, Gao D, Wang L, Liu S, Zhao J, Li X, Qian P (2023) The multidrug-resistant Pseudomonas fluorescens strain: a hidden threat in boar semen preservation. Front Microbiol 14:1279630

    Article  PubMed  PubMed Central  Google Scholar 

  38. Schubert B, Badiou M, Force A (2019) Computer-aided sperm analysis, the new key player in routine sperm assessment. Andrologia 51:e13417

    Article  PubMed  Google Scholar 

  39. Fraser L, Brym P, Pareek CS, Mogielnicka-Brzozowska M, Paukszto Ł, Jastrzębski JP, Wasilewska-Sakowska K, Mańkowska A, Sobiech P, Żukowski K (2020) Transcriptome analysis of boar spermatozoa with different freezability using RNA-Seq. Theriogenology 142:400–413

    Article  PubMed  CAS  Google Scholar 

  40. Santiani A, Juárez J, Allauca P, Roman B, Ugarelli A, Evangelista-Vargas S (2023) Cryopreservation effect on sperm viability, mitochondrial membrane potential, acrosome integrity and sperm capacitation of alpaca spermatozoa detected by imaging flow cytometry. Reprod Domest Anim 58:560–563

    Article  PubMed  CAS  Google Scholar 

  41. Díaz-Bertrana ML, Deleuze S, Pitti Rios L, Yeste M, Morales Fariña I, Rivera Del Alamo MM (2021) Microbial prevalence and antimicrobial sensitivity in equine endometritis in field conditions. Animals (Basel) 11:1476

    Article  PubMed  Google Scholar 

  42. Xia F, Li X, Wang B, Gong P, Xiao F, Yang M, Zhang L, Song J, Hu L, Cheng M, Sun C, Feng X, Lei L, Ouyang S, Liu ZJ, Li X, Gu J, Han W (2016) Combination therapy of LysGH15 and apigenin as a new strategy for treating pneumonia caused by Staphylococcus aureus. Appl Environ Microbiol 82:87–94

    Article  PubMed  CAS  Google Scholar 

  43. Li X, Wang S, Nyaruaba R, Liu H, Yang H, Wei H (2021) A highly active chimeric lysin with a calcium-enhanced bactericidal activity against Staphylococcus aureus in vitro and in vivo. Antibiotics (Basel) 10:461

    Article  PubMed  CAS  Google Scholar 

  44. Naranjo-Lucena A, Slowey R (2023) Invited review: Antimicrobial resistance in bovine mastitis pathogens: A review of genetic determinants and prevalence of resistance in European countries. J Dairy Sci 106:1–23

    Article  PubMed  CAS  Google Scholar 

  45. Pitkälä A, Haveri M, Pyörälä S, Myllys V, Honkanen-Buzalski T (2004) Bovine mastitis in Finland 2001–prevalence, distribution of bacteria, and antimicrobial resistance. J Dairy Sci 87:2433–2441

    Article  PubMed  Google Scholar 

  46. Ruegg PL (2017) A 100-year review: mastitis detection, management, and prevention. J Dairy Sci 100:10381–10397

    Article  PubMed  CAS  Google Scholar 

  47. Gomes F, Saavedra MJ, Henriques M (2016) Bovine mastitis disease/pathogenicity: evidence of the potential role of microbial biofilms. Pathog Dis 74:ftw006

    Article  PubMed  Google Scholar 

  48. Wilson DN (2014) Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat Rev Microbiol 12:35–48

    Article  PubMed  CAS  Google Scholar 

  49. Gutiérrez D, Fernández L, Rodríguez A, García P (2018) Are phage lytic proteins the secret weapon to kill Staphylococcus aureus? MBio 9:e01923-17

    Article  PubMed  PubMed Central  Google Scholar 

  50. Maes D, Nauwynck H, Rijsselaere T, Mateusen B, Vyt P, de Kruif A, Van Soom A (2008) Diseases in swine transmitted by artificial insemination: an overview. Theriogenology 70:1337–1345

    Article  PubMed  CAS  Google Scholar 

  51. Chen Y, Li X, Wang S, Guan L, Li X, Hu D, Gao D, Song J, Chen H, Qian P (2020) A novel tail-associated O91-specific polysaccharide depolymerase from a podophage reveals lytic efficacy of Shiga toxin-producing Escherichia coli. Appl Environ Microbiol 86:e00145-e220

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Cicinelli E, De Ziegler D, Nicoletti R, Colafiglio G, Saliani N, Resta L, Rizzi D, De Vito D (2008) Chronic endometritis: correlation among hysteroscopic, histologic, and bacteriologic findings in a prospective trial with 2190 consecutive office hysteroscopies. Fertil Steril 89:677–684

    Article  PubMed  Google Scholar 

  53. Kitaya K, Matsubayashi H, Takaya Y, Nishiyama R, Yamaguchi K, Takeuchi T, Ishikawa T (2017) Live birth rate following oral antibiotic treatment for chronic endometritis in infertile women with repeated implantation failure. Am J Reprod Immunol 78:6

    Article  Google Scholar 

  54. Cimadomo D, Craciunas L, Vermeulen N, Vomstein K, Toth B (2021) Definition, diagnostic and therapeutic options in recurrent implantation failure: an international survey of clinicians and embryologists. Hum Reprod 36:305–317

    Article  PubMed  CAS  Google Scholar 

  55. Coughlan C, Ledger W, Wang Q, Liu F, Demirol A, Gurgan T, Cutting R, Ong K, Sallam H, Li TC (2014) Recurrent implantation failure: definition and management. Reprod Biomed Online 28:14–38

    Article  PubMed  CAS  Google Scholar 

  56. Ballas P, Pothmann H, Pothmann I, Drillich M, Ehling-Schulz M, Wagener K (2022) Dynamics and diversity of intrauterine anaerobic microbiota in dairy cows with clinical and subclinical endometritis. Animals (Basel) 13:82

    Article  PubMed  Google Scholar 

  57. WHO (2019) Critically important antimicrobials for human medicine: 6th revision. https://www.who.int/publications/i/item/9789241515528. Accessed 26 Oct 2024

  58. Hao H, Pan H, Ahmad I, Cheng G, Wang Y, Dai M, Tao Y, Chen D, Peng D, Liu Z, Huang L, Yuan Z (2013) Susceptibility breakpoint for enrofloxacin against swine Salmonella spp. J Clin Microbiol 51:3070–3072

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Zhu H, Yao J, Zhang Z, Jiang X, Zhou Y, Bai Y, Hu X, Ning H, Hu J (2022) Sulfidised nanoscale zerovalent iron-modified pitaya peel-derived carbon for enrofloxacin degradation and swine wastewater treatment: Combination of electro-Fenton and bio-electro-Fenton process. J Hazard Mater 434:128767

    Article  PubMed  CAS  Google Scholar 

  60. Zhang L, Wu X, Huang Z, Zhang N, Wu Y, Cai Q, Shen X, Ding H (2018) Pharmacokinetic/pharmacodynamic assessment of cefquinome against Actinobacillus Pleuropneumoniae in a piglet tissue cage infection model. Vet Microbiol 219:100–106

    Article  PubMed  Google Scholar 

  61. Wang Z, Ma J, Wang J, Yang D, Kong L, Fu Q, Cheng Y, Wang H, Yan Y, Sun J (2019) Application of the phage lysin Ply5218 in the treatment of Streptococcus suis infection in piglets. Viruses 11:82

    Article  Google Scholar 

  62. Lv M, Wang S, Yan G, Sun C, Feng X, Gu J, Han W, Lei L (2015) Genome sequencing and analysis of an Escherichia coli phage vB_EcoM-ep3 with a novel lysin, Lysep3. Virus Genes 50:487–497

    Article  PubMed  CAS  Google Scholar 

  63. Larpin Y, Oechslin F, Moreillon P, Resch G, Entenza JM, Mancini S (2018) In vitro characterization of PlyE146, a novel phage lysin that targets Gram-negative bacteria. PLoS One 13:e0192507

    Article  PubMed  PubMed Central  Google Scholar 

  64. Nehru DA, Dhaliwal GS, Jan MH, Cheema RS, Kumar S (2019) Clinical efficacy of intrauterine cephapirin benzathine administration on clearance of uterine bacteria and subclinical endometritis in postpartum buffaloes. Reprod Domest Anim 54:317–324

    Article  PubMed  CAS  Google Scholar 

  65. Cicinelli E, Matteo M, Trojano G, Mitola PC, Tinelli R, Vitagliano A, Crupano FM, Lepera A, Miragliotta G, Resta L (2018) Chronic endometritis in patients with unexplained infertility: Prevalence and effects of antibiotic treatment on spontaneous conception. Am J Reprod Immunol 79:e12782

    Article  Google Scholar 

  66. Wire MB, Jun SY, Jang IJ, Lee SH, Hwang JG, Huang DB (2022) A phase 1 study to evaluate safety and pharmacokinetics following administration of single and multiple doses of the antistaphylococcal lysin LSVT-1701 in healthy adult subjects. Antimicrob Agents Chemother 66:e0184221

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Home for Researchers editorial team for language editing services. We thank the National Key Laboratory of Agricultural Microbiology Core Facility, Huazhong Agricultural University, for assisting in the expression and purification of ClyL and Lys0859.

Funding

This work was supported by grants from the Agricultural Science and Technology Research project of Hubei Province [HBNYHXGG2023-3], the National Program on Key Research Project of China [2022YFD1800800], “Yingzi Tech & Huazhong Agricultural University Intelligent Research Institute of Food Health” [No. IRIFH202209; IRIFH202301], and the National Key Laboratory of Agricultural Microbiology [AML2023B11].

Author information

Authors and Affiliations

Authors

Contributions

XXL drafted the main manuscript, and XXL and ZH performed the data analysis; XXL, ZH, ZX, LZ, SW, and PC planned and performed the experiments; PT, X-ML, and PQ were responsible for the experimental design and funding. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Pin Chen, Xiang-min Li or Ping Qian.

Ethics declarations

Ethics approval and consent to participate

All animal experiments were approved by the Laboratory Animal Monitoring Committee of Huazhong Agricultural University and carried out in accordance with the corresponding guidelines for laboratory animal operations at Huazhong Agricultural University. The ethical approval codes for mouse and swine are 202311160010 and YFXJ-2023001, respectively.

Competing interests

The authors declare that they have no competing interests.

Additional information

Handling editor: Marcelo Gottschalk

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1. Primers used to distinguish bacteria from sow endometritis.

Additional file 2. Primers used to identify the serotypes of

S. suis.

Additional file 3. Criteria used to determine the antibiotic sensitivity of bacteria from sows with endometritis.

Additional file 4. Primers used to construct the chimeric lysin ClyL.

Additional file 5. MIC distributions of fourteen antibiotics against

Staphylococcus spp. (n = 141).

Additional file 6. MIC distributions of fourteen antibiotics to

S. suis (n = 66).

Additional file 7. MIC distributions of eleven antibiotics against

Escherichia coli (n = 155).

Additional file 8. Turbidity decreases in the effects of the phage lysins ClyL and Lys0859 against

S. suis.

Additional file 9. Turbidity decrease caused by the phage lysins ClyL and Lys0859 against

Staphylococcus.

Additional file 10. FIC indices for the combinations of ClyL and Lys0859.

Additional file 11. Chimeric lysin ClyL and parental phage lysin Lys0859-treated 24-h- and 48-h-old biofilms formed from single

S. aureus ATCC29213 and S. agalactiae ATCC13813 strains. The biomass of single S. aureus ATCC29213 (A) and S. agalactiae ATCC13813 (B) biofilms was treated with ClyL and Lys0859 at 37 °C for 2 h. The viable cell counts of single S. aureus ATCC29213 (C) and S. agalactiae ATCC13813 (D) biofilms were determined after treatment with ClyL and Lys0859 at 37 °C for 2 h. Significant differences between the PBS groups and the ClyL and Lys0859 groups were determined by Student’s t test (ns p > 0.05, * p < 0.05 ** p < 0.01, and *** p < 0.001). The experiments were performed three times. The data are shown as the mean ± SD.

Additional file 12. Effects of ClyL, Lys0859 and cefquinome on various kinematic parameters (motility) of sperm.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Xx., Hong, Zq., Xiong, Zx. et al. Development of a novel chimeric lysin to combine parental phage lysin and cefquinome for preventing sow endometritis after artificial insemination. Vet Res 56, 39 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13567-025-01457-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13567-025-01457-4

Keywords