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

Bacteriophages isolated from dairy farm mitigated Klebsiella pneumoniae-induced inflammation in bovine mammary epithelial cells cultured in vitro

Abstract

Background

Klebsiella pneumoniae, an environmental pathogen causing mastitis in dairy cattle, is often resistant to antibiotics. K. pneumoniae was used as the host bacteria to support bacteriophage replication; 2 bacteriophages, CM8-1 and SJT-2 were isolated and considered to have therapeutic potential. In the present study, we determined the ability of these 2 bacteriophages to mitigate cytotoxicity, pathomorphological changes, inflammatory responses and apoptosis induced by K. pneumoniae (bacteriophage to K. pneumoniae MOI 1:10) in bovine mammary epithelial cells (bMECs) cultured in vitro.

Results

Bacteriophages reduced bacterial adhesion and invasion and cytotoxicity (lactate dehydrogenase release). Morphological changes in bMECs, including swelling, shrinkage, necrosis and hematoxylin and eosin staining of cytoplasm, were apparent 4 to 8 h after infection with K. pneumoniae, but each bacteriophage significantly suppressed damage and decreased TNF-α and IL-1β concentrations. K. pneumoniae enhanced mRNA expression of TLR4, NF-κB, TNF-α, IL-1β, IL-6, IL-8, caspase-3, caspase-9 and cyt-c in bMECs and increased apoptosis of bMECs, although these effects were mitigated by treatment with either bacteriophage for 8 h.

Conclusions

Bacteriophages CM8-1 and SJT-2 mitigated K. pneumoniae-induced inflammation in bMECs cultured in vitro. Therefore, the potential of these bacteriophages for treating mastitis in cows should be determined in clinical trials.

Background

Bovine mastitis caused by infection with pathogenic microorganisms reduces milk production and quality, inflicting huge economic losses in the dairy industry [1, 2]. Klebsiella pneumoniae, a common environmental pathogen, causes mastitis in dairy cows [3, 4], accounting for 10 to 13% clinical mastitis, second to E. coli [5, 6]. Although antimicrobials are the most common therapy for bovine mastitis caused by K. pneumoniae, it often has single or multidrug resistance [4], with increasing risk of recurrence after treatment.

Bovine mammary epithelial cells (bMECs) are the first line of defense against invading pathogens that enter the udder through the streak canal, at the distal end of the bovine teat [4, 7]. Thus, bMECs have critical roles in both nonspecific and specific immune defenses against infection by pathogenic microorganisms [7]; furthermore, they can be infected by a variety of pathogenic microorganisms that damage these cells, leading to mastitis [8, 9]. Toll-like Receptor 4 (TLR4) and the NF-κB pathway are classic regulators of inflammation [10]. Lipopolysaccharides (LPS), the main component of the cell wall of gram-negative bacteria, can activate the TLR4-mediated NF-κB pathway to induce an inflammatory response [11]. Inhibiting or knocking out TLR4 down-regulated expression of inflammatory factors in cells, thereby suppressing inflammation [11, 12]. Although a timely and appropriate inflammatory response can effectively contain invading pathogens, an excessive inflammatory response can cause various degrees of cellular damage, including apoptosis, necrosis or pyroptosis [13, 14]. K. pneumoniae can multiply rapidly in bovine mammary tissues, causing a severe inflammatory response and tissue damage [4, 15]. In bovine mastitis induced by K. pneumoniae, infection of bMECs is an important component. Antibiotics are commonly used to treat bovine mastitis and consequently, drug resistance in gram-negative bacteria, including K. pneumoniae, is rapidly increasing [3, 16, 17]. Therefore, it is important to identify alternatives to antibiotics for treating bacterial infections, especially for bovine mastitis caused by K. pneumoniae.

Bacteriophages are small viruses, nearly ubiquitous in the natural environment [18]. Bacteriophage can lyse pathogens in vivo and in vitro without harmful effects on host cells and have been widely used in various fields [19, 20]. For example, 3 Myoviridae bacteriophages were used as adjunctive therapy and were safe and effective with severe Staphylococcus aureus infections [21]. Furthermore, a bacteriophage “cocktail” was very successful in controlling bacterial wilt in tomatoes [22]. However, use of a bacteriophage for treatment of bovine mastitis caused by K. pneumoniae has apparently not been reported. Therefore, our objective was to investigate the role of bacteriophages for controlling K. pneumoniae in an in vitro infection model of bMECs.

Results

Transmission electron microscopy (TEM) of bacteriophages

Ultrastructures of K. pneumoniae bacteriophages CM8-1 and SJT-2 are shown (Fig. 1). Bacteriophages CM8-1 and SJT-2 were comprised of a head, neck and tail wire (Fig. 1A2, B2). Bacteriophages CM8-1 and SJT-2 had a polyhedral head, approximately 100 and 103 nm, respectively, whereas their tails were 103 and 116 nm and contained obvious tail filaments (Fig. 1A2, B2).

Fig. 1
figure 1

Transmission electron micrographs of Klebsiella pneumoniae bacteriophages CM8-1 and SJT-2. A1 and A2 are the ultrastructure of phage CM8-1, whereas B1 and B2 are the ultrastructure of phage SJT-2. Black arrows indicate tail filaments

Effect of bacteriophages on adhesion and invasion of K. pneumoniae

Adhesion of K. pneumoniae to bMECs in the K. pneumoniae, K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were significantly increased in comparison to the Control (Fig. 2a). Starting soon after K. pneumoniae infection and as interval after infection increased, bacteriophages CM8-1 and SJT-2 reduced adhesion (P < 0.01) of K. pneumoniae to bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups as compared to the K. pneumoniae group at 1.5, 2, 2.5 and 3 h post infection (hpi) (Fig. 2a).

Fig. 2
figure 2

Adhesion and invasion of Klebsiella pneumoniae-infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. a Adhesion of K. pneumoniae to bMECs, b Invasion of K. pneumoniae into bMECs. Compared to the previous time point in the same group: aP < 0.01; compared to the Control group: *P < 0.05, **P < 0.01; compared to the K. pneumoniae group: #P < 0.05, ##P < 0.01

Adhesion of K. pneumoniae to bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was similar to that in the K. pneumoniae group, without significant changes at 0.5 hpi (Fig. 2B). However, as duration of infection increased, adhesion of K. pneumoniae to bMECs in the K. pneumoniae, K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups increased (P < 0.01) compared to the Control group (Fig. 2B). Furthermore, adhesion of K. pneumoniae to bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was lower (P < 0.01) than that of the K. pneumoniae group at 2, 2.5 and 3 hpi (Fig. 2B).

Effect of bacteriophages on K. pneumoniae-induced cytotoxicity in bMECs

There was no difference in LDH release among the Control, CM8-1 and SJT-2 groups (Fig. 3). At 2 hpi with K. pneumoniae, LDH release in the K. pneumoniae group was higher (P < 0.01) than in the Control group (Fig. 3), whereas at 4, 6 and 8 hpi, LDH release in the K. pneumoniae, K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was greater (P < 0.01) than in the Control group (Fig. 3). At 2, 4, 6 and 8 hpi, LDH release in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was significantly decreased compared to the K. pneumoniae group (Fig. 3).

Fig. 3
figure 3

Lactate dehydrogenase (LDH) release from Klebsiella pneumoniae-infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. Compared to the Control group: *P < 0.05, **P < 0.01; compared to the K. pneumoniae group: #P < 0.05, ##P < 0.01

Fig. 4
figure 4

Morphological observations (40×) of Klebsiella pneumoniae-infected bMECs in presence of bacteriophages CM8-1 or SJT-2. A1-D1: Normal bMECs in the Control group at 2, 4, 6 and 8 h. A2-D2: Note damage to bMECs, including necrosis and swelling, shrinkage, loss of cell integrity, necrosis and heavy staining, and reductions in cell number at 2, 4, 6 and 8 h after infection in K. pneumoniae group. A3-D3 and A4-D4: Phages CM8-1 and SJT-2 mitigated cell damage caused by K. pneumoniae infection at 2, 4, 6 and 8 h in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups, respectively

Morphological changes in bMECs

At 2 hpi with K. pneumoniae, morphological appearance was similar in the K. pneumoniae and Control groups. At 4 and 6 hpi with K. pneumoniae, there was no significant changes compared to the Control group (Fig. 4A1-C1, A2-C2). However, at 8 hpi with K. pneumoniae, damage to bMECs in K. pneumoniae group included cell swelling, nuclear hyperchromatism and necrosis, as well as reductions in number of bMECs, compared to the Control group (Fig. 4D1-D2). Furthermore, the extent of damage to bMECs gradually increased with interval after infection. However, morphology of bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was similar compared to the K. pneumoniae and Control groups at 2 hpi. Swelling and necrosis of bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were less severe compared to the K. pneumoniae group at 4, 6 and 8 hpi (Fig. 4A3-D3, A4-D4).

Effect of bacteriophages on concentrations of inflammatory factors and inflammatory responses in K. pneumoniae-infected bMECs

Concentrations of IL-1β and TNF-α proteins were increased (P < 0.01) at 4, 6 and 8 hpi with K. pneumoniae (Fig. 5a and b). However, TNF-α and IL-1β concentrations in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were significantly decreased in comparison to the K. pneumoniae group at 4, 6 and 8 hpi (Fig. 5a and b).

Fig. 5
figure 5

Concentrations of IL-1β and TNF-α proteins in the supernatant of Klebsiella pneumoniae-infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. a Content of IL-1β in the supernatant of K. pneumoniae infected bMECs, b Content of TNF-α in the supernatant of K. pneumoniae infected bMECs. Data presented as mean ± SD for 3 independent experiments. Compared to the Control group: *P < 0.05, **P < 0.01; compared to the K. pneumoniae group: #P < 0.05, ##P < 0.01

Expression levels of TLR4, NF-κB and inflammatory factors (TNF-α, IL-1β, IL-6 and IL-8) in bMECs were determined by real-time PCR. At 2 hpi, mRNA expression levels of TLR4, NF-κB, TNF-α, IL-1β, IL-6 and IL-8 in bMECs were significantly higher in the K. pneumoniae versus Control groups (Fig. 6a-f). Furthermore, at 4, 6 and 8 hpi, mRNA expression levels of TLR4, NF-κB and inflammatory factors in the K. pneumoniae, K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were significantly up-regulated compared to the Control group (Fig. 6a-f). In contrast, after treatment with bacteriophages CM8-1 and SJT-2, at 4, 6 and 8 hpi, mRNA expression levels of TLR4, NF-κB and inflammatory factors in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were significantly down-regulated as compared to the K. pneumoniae group (Fig. 6a-f).

Fig. 6
figure 6

Relative mRNA expressions of TLR 4, NF-κB, IL-1β, TNF-α, IL-6 and IL-8 in Klebsiella pneumonia infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. Data presented as mean ± SD for 3 independent experiments. Compared to the Control group: *P < 0.05, **P < 0.01; compared to the K. pneumoniae group: #P < 0.05, ##P < 0.01

Effect of bacteriophages on apoptosis in infected bMECs

At 2 hpi of bMECs with K. pneumoniae, mRNA expression levels of apoptosis factors caspase-3 and caspase-9 were higher (P < 0.05) in the K. pneumoniae groups than in the Control group (Fig. 7a, b). At 4, 6 and 8 hpi, mRNA expression levels of apoptosis factors in the K. pneumoniae, K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups were significantly up-regulated compared to Controls (Fig. 7a-c). When compared to the K. pneumoniae group, at 4, 6 and 8 hpi, bacteriophages CM8-1 and SJT-2 significantly down-regulated mRNA expression levels of apoptosis factors in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups (Fig. 7a-c).

Fig. 7
figure 7

Relative mRNA expressions of apoptosis-related genes caspase 3, caspase 9 and cyt c in Klebsiella pneumoniae-infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. Data presented as mean ± SD for 3 independent experiments. Compared to the Control group: *P < 0.05, **P < 0.01; compared to the K. pneumoniae group: #P < 0.05, ##P < 0.01

Apoptosis of bMECs in the K. pneumoniae group was higher (P < 0.05) than in the Control group at 2 hpi (Fig. 8). Furthermore, apoptosis rate of bMECs in the K. pneumoniae + CM8-1 and K. pneumoniae + SJT-2 groups was lower (P < 0.01) compared to the K. pneumoniae group at 4, 6 and 8 hpi (Fig. 8), although apoptosis was significantly higher in each of these groups compared to the Control group at all of these time points (Fig. 8).

Fig. 8
figure 8

Flow cytometry to assess apoptosis rate by of Klebsiella pneumoniae-infected bovine mammary epithelial cells in presence of bacteriophages CM8-1 or SJT-2. A-D Percentage of apoptotic cells stained with FITC-conjugated annexin-V and PI (up to 8 hpi). E Percentage of apoptotic cells. Data are mean ± SD for 3 independent experiments. Compared to the Control group: *P < 0.05, **P < 0.01; compared to K. pneumoniae group: #P < 0.05, ##P < 0.01

Discussion

This was apparently the first report that bacteriophages CM8-1 and SJT-2 isolated from mastitic bovine milk mitigated inflammatory responses and apoptosis in bMECs induced by K. pneumoniae by decreasing bacterial adhesion and invasion and suppressing expression of inflammatory factors. We concluded that bacteriophages CM8-1 and SJT-2 were protective in K. pneumoniae-infected bMECs and therefore have potential therapeutic value against bovine mastitis caused by K. pneumoniae.

K. pneumoniae can infect a variety of cells and tissues, inducing inflammatory responses and cell damage [23,24,25]. In addition, it also infects bMECs in development of mastitis in dairy cows [26]. When bacteria infect cells, bacterial adhesion and invasion are critical determinants of virulence [27]; therefore, the absence of genes related to synthesis of adhesion proteins reduce adhesion rates and cell toxicity. Furthermore, pathogens with high rates of adhesion and invasion have potential to cause serious damage to cells. In this study, adhesion and invasion of K. pneumoniae to bMECs increased significantly with interval after infection, although this was mitigated by bacteriophages CM8-1 and SJT-2. Bacteriophages can specifically lyse pathogens and therefore reduce adhesion and invasion. In the current study, bacteriophages CM8-1 and SJT-2 reduced ability of K. pneumoniae to infect and bMECs by mitigating adhesion and invasion.

Lactate dehydrogenase (LDH) is an important marker of bacterial cytotoxicity [28, 29]. Infection of bMECs with gram-negative bacteria, including K. pneumoniae and E. coli, increases LDH release and is accompanied by substantial cell damage [4, 30]. In this study, LDH significantly increased after bMECs were infected by K. pneumoniae, with increasing concentrations over time. Therefore, K. pneumoniae had toxic effects on bMECs, consistent with cell swelling and necrosis and decreasing numbers of bMECs.

A lack of toxic effects on host cells is characteristic of bacteriophages [19, 31]. Treatment of bMECs with either CM8-1 or SJT-2 bacteriophage did not increase release of LDH relative to the Control group, indicating that these bacteriophages were not toxic to bMECs. However, they can lyse and kill K. pneumoniae, reducing its virulence and mitigating damage to bMECs, manifested by less morphological damage and lower LDH concentrations.

Infection of bMECs by pathogens in development of bovine mastitis is usually accompanied by a severe inflammatory response [32]. Various pathogenic bacteria, including K. pneumoniae, E. coli, S. aureus and S. agalactiae, cause inflammation by producing inflammatory factors [4, 33,34,35]. In this study, concentrations of IL-1β and TNF-α in bMECs culture medium increased significantly after bMECs were infected with K. pneumoniae; furthermore, there was a positive association with duration of infection time, confirming K. pneumoniae-induced inflammation of bMECs. Gram negative bacteria such as K. pneumoniae and E. coli can activate TLR4 mediated NF-κB pathway to induce inflammation of bMECs [4, 11, 36, 37]. In this study, after exposing bMECs to K. pneumoniae, mRNA levels of TLR4 and NF-κB, in the pathway of regulating inflammatory response in bMECs, were significantly up-regulated and positively associated with duration of infection. In addition, downstream genes in TLR4 and NF-κB pathways, namely mRNA expression levels of TNF-α, IL-1β, IL-6 and IL-8 in bMECs were also up-regulated, confirming that K. pneumoniae induced an inflammatory response in bMECs. However, bacteriophages CM8-1 and SJT-2 were able to lyse K. pneumoniae and mitigate the inflammatory response induced by K. pneumoniae, manifested by decreased concentrations of IL-1β and TNF-α in bMECs culture medium, as well as reduced mRNA expression levels of TLR4, NF-κB, TNF-α, IL-1β, IL-6 and IL-8.

In this study, K. pneumoniae induced apoptosis of bMECs. Mitochondria are involved in the common apoptotic pathway, which mainly depends on the caspase families and cyt-c for regulation [38,39,40]. In the current study, K. pneumoniae infection of bMECs presumably enhanced release of cyt-c from mitochondria, triggered the caspase-9 and caspase-3 cascade reaction and accelerated cell apoptosis. The mRNA expression of caspase-3, caspase-9 and cyt-c in bMECs significantly increased with duration of infection time, implying apoptosis was triggered by K. pneumoniae. Based on flow cytometry, the apoptosis rate of bMECs increased gradually after K. pneumoniae treatment, which further confirmed that K. pneumoniae induced apoptosis of bMECs. However, bacteriophages CM8-1 and SJT-2 decreased expression of caspase-3, caspase-9 and cyt-c, as well as apoptosis rate of bMECs. Therefore, bacteriophages CM8-1 and SJT-2 mitigated apoptosis of bMECs induced by K. pneumoniae.

Conclusions

Bacteriophages CM8-1 and SJT-2 mitigated K. pneumoniae-induced inflammation in bovine mammary epithelial cells cultured in vitro. Therefore, the potential of these bacteriophages for treating mastitis in cows should be determined in clinical trials.

Methods

Bacterial strains

The bacterial strain used in this study, isolated from clinical mastitis in a dairy cow, was K. pneumoniae (CM-KP8) with K57 capsule serotype [4]. This isolate was stored at the mastitis diagnostic laboratory, College of Veterinary Medicine, China Agricultural University. Prior to each experiment, fresh bacterial suspensions were prepared from frozen stocks by culture on LB nutrient agar medium and incubated at 37 oC for 18 h. Thereafter, bacteria were sub-cultured on brain heart infusion broth for 18 h to mid-log phase and bacterial concentration was determined by the plate counting method for the following experiments.

Bacteriophages

Bacteriophages CM8-1 and SJT-2 were isolated from dairy farm wastewater. Prior to each experiment, a bacterial solution in mid-log phase was mixed with bacteriophage stock solution and spread over a double-layer plate (96.6% LB liquid medium and 3.4% agar powder), with sodium magnesium (SM) buffer added after the bacteriophage had grown on the entire plate. Thereafter, this plate was placed on a shaker at 120 rpm/min for 2 h to liberate the bacteriophage from the plate and into the SM solution. The latter was collected and passed through a 0.22 µm filter to obtain the bacteriophage stock solution. Thereafter, PEG8000 (10%) was add into the bacteriophage suspension and left overnight at 4 oC. Then, the bacteriophage suspension was centrifuged at 10,000 × g for 10 min to obtain bacteriophage precipitation. The above process was repeated twice to obtain pure bacteriophage. The bacteriophage precipitation was resuspended with DMEM for the following experiments.

Cell culture and treatment

Bovine mammary epithelial cells (bMECs), line MAC-T (Shanghai Jingma Biological Technology Co., Ltd. China), were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% (v/v) Gibco® fetal bovine serum at 37 oC in a humidified atmosphere of 5% CO2. Cells from 3 to 8 passages were used for experiments, with the same passages of cells used in each experiment. The bMECs were treated with or without K. pneumoniae (MOI, ratio of K. pneumoniae to bMECs was 10:1) and bacteriophages CM8-1 or SJT-2 (MOI, ratio of bacteriophage to K. pneumoniae was 1:10), to form the following groups: Control; K. pneumoniae infection group (K. pneumoniae), K. pneumoniae plus bacteriophage CM8-1 group (K. pneumoniae + CM8-1) and K. pneumoniae plus bacteriophage SJT-2 group (K. pneumoniae + SJT-2).

Transmission electron microscopy (TEM)

A suspension (20 µL) of bacteriophages (titer = 109 PFU/mL) was dropped on a copper mesh and precipitated for 2 min at room temperature, with excess liquid absorbed by filter paper. Bacteriophages were then stained with 2% uranium acetate for 1 min, excess liquid absorbed, naturally dried for 15 min at room temperature and then observed under a transmission electron microscope (H-7650, Hitachi, Tokyo, Japan) at 80 kV.

Adhesion and invasion

Adhesion and invasion were measured as described [4]. The bMECs were inoculated into a 6-well cell culture plate and grown to 80% confluence, washed with phosphate buffer saline (PBS) and then DMEM supplemented with 1% (v/v) Gibco® Fetal Bovine Serum was added. At a multiplicity of infection (MOI, ratio of K. pneumoniae to cells) of 10:1, the bMECs were cultured at 37 °C in a humidified atmosphere of 5% CO2 for 0.5 h, and then with addition of bacteriophages CM8-1 or SJT-2 (MOI, ratio of bacteriophages to K. pneumoniae) of 1:10 for 0.5, 1, 1.5, 2 and 2.5 h. After incubation, cells were washed with PBS (to remove non-adherent bacteria), and the cell suspension was 10-fold diluted and cultured on LB liquid medium plate for counting colony forming units. Meanwhile, after incubation, cells were washed with PBS and treated with 1 mL of DMEM containing kanamycin (100 µg/mL) and then cultured at 37 °C in a humidified atmosphere of 5% CO2 for 2 h. The culture medium was removed, cells were lysed with 0.5% TritonX-100 and the lysate diluted and cultured on LB liquid medium plate as described above. Colonies were counted and invasion rate of K. pneumoniae into bMECs was calculated.

Lactate dehydrogenase (LDH) assay

The bMECs were seeded in 96-well plates (density of 1 × 105 cells/well), grown to 80% confluence, then DMEM supplemented with 1% (v/v) Gibco® Fetal Bovine Serum was added. Cells were then treated with K. pneumoniae and bacteriophages CM8-1 or SJT-2, as described above. After culture for 1, 2, 4, 6 or 8 h, supernatants were collected, centrifuged at 11,586 × g for 15 min at 4 oC and LDH release measured by cytotoxicity (LDH Assay Kit-WST®, Dojingdo Laboratories, Kumamoto, Japan), according to manufacturer’s instructions. Absorbance of optical density was determined at 492 nm with a microplate reader (Bio-Rad, Hercules, CA, USA).

Morphological observation of bMECs

The bMECs were seeded in 6-well plates with glass slides and grown to 80% confluence. Cells were then treated with K. pneumoniae and bacteriophages CM8-1 or SJT-2, as described above. Cells were cultured at 37 °C in a humidified atmosphere of 5% CO2 for 2, 4, 6 or 8 h. Then, cells were collected, fixed in 4% paraformaldehyde for 30 min, washed 3 times with PBS and dried naturally for 5 min. Hematoxylin-eosin (HE) staining was performed according to manufacturer’s instructions. An increasing alcohol gradient (75, 85, 95 and 100%) was used to dehydrate cells, which were subsequently observed under a light microscope at 40×.

Detection of cytokine concentrations

Culture medium was collected, centrifuged at 11,586 × g for 10 min at 4 oC and supernatant removed. Concentrations of inflammatory cytokines TNF-α and IL-1β in supernatant were measured using ELISA kits (Shanghai Zhenke Biotechnology Co., Ltd, Shanghai, China), used in accordance with manufacturer’s instructions.

Quantifying cytokine mRNA with real‐time PCR

The bMECs were treated with K. pneumoniae and bacteriophages CM8-1 or SJT-2, and cells collected for total RNA extraction. Trizol Reagent (1 mL) was pre-chilled on ice and added to cell samples for 5 min to lyse cells. Mixed liquid was centrifuged at 11,586 × g for 15 min at 4 oC and supernatant collected. Total mRNA of bMECs was extracted with mRNA extraction kit (TransGen Biotech Co., Ltd. Beijing, China), used according to manufacturer’s instructions. Primers for GAPDH (housekeeping gene), TLR4, NF-κB, TNF-α, IL-1β, IL-6, IL-8, caspase-3, caspase-9 and cyt-c were designed using primer 5.0 software (Table 1). Relative expression levels of TLR4, NF-κB, TNF-α, IL-1β, IL-6, IL-8, caspase-3, caspase-9 and cyt-c mRNA were determined using the Mx3000P™ RT-PCR system (Stratagene, USA) real-time PCR RG-3000A and the SYBR® Premix Ex Taq™ (Perfect Real Time) Kit (Takara, Japan).

Table 1 List of primers for real-time PCRa

Detection of apoptosis by flow cytometry

After bMECs were treated with K. pneumoniae and bacteriophages CM8-1 and SJT-2, apoptosis was assessed with an Annexin V-FITC apoptosis detection kit (Beyotime Biotechnology Co., Ltd, Shanghai, China). Cell culture fluid and washing fluid were collected. Then, cells were harvested with trypsin without EDTA and washed with PBS. Cells were centrifuged at 965 × g for 10 min, supernatant removed and cells re-suspended in 195 µL Annexin V-FITC binding solution and then transferred to a sterile flow cytometry glass tube. Annexin V-FITC (5 µL) was add to the cell suspension, followed by 10 µL propidium iodide staining solution. After 20 min at room temperature in dark conditions, apoptotic cells were detected using flow cytometry.

Statistical analyses

Data were expressed as mean ± standard deviation (SD), followed by post hoc Tukey’s tests using SPSS 22.0 (SPSS Inc., Chicago, IL, USA). All measurements were replicated three times and P < 0.05 was considered significant.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

K. pneumoniae :

Klebsiella pneumoniae

LDH:

Lactate dehydrogenase

bMECs:

bovine mammary epithelial cells

MOI:

Multiplicity of infection

TLR:

Toll-like Receptor

SM:

Sodium magnesium

DMEM:

Dulbecco’s modified Eagle’s medium

HE:

Hematoxylin-eosin

TEM:

Transmission electron microscopy

PCR:

Polymerase chain reaction

References

  1. Heikkilä AM, Liski E, Pyörälä S, Taponen S. Pathogen-specific production losses in bovine mastitis. J Dairy Sci. 2018;101:9493–504.

    Article  Google Scholar 

  2. He W, Ma S, Lei L, He J, Li X, Tao J, Wang X, Song S, Wang Y, Wang Y, Shen J, Cai C, Wu C. Prevalence, etiology, and economic impact of clinical mastitis on large dairy farms in China. Vet Microbiol. 2020;242:108570.

    Article  Google Scholar 

  3. Cheng J, Qu W, Barkema HW, Nobrega DB, Gao J, Liu G, De Buck J, Kastelic JP, Sun H, Han B. Antimicrobial resistance profiles of 5 common bovine mastitis pathogens in large Chinese dairy herds. J Dairy Sci. 2019;102:2416–26.

    Article  CAS  Google Scholar 

  4. Cheng J, Zhang J, Han B, Barkema HW, Cobo ER, Kastelic JP, Zhou M, Shi Y, Wang J, Yang R, Gao J. Klebsiella pneumoniae isolated from bovine mastitis is cytopathogenic for bovine mammary epithelial cells. J Dairy Sci. 2020;103:3493–504.

    Article  CAS  Google Scholar 

  5. Gao J, Barkema HW, Zhang L, Liu G, Deng Z, Cai L, Shan R, Zhang S, Zou J, Kastelic JP, Han B. Incidence of clinical mastitis and distribution of pathogens on large Chinese dairy farms. J Dairy Sci. 2017;100:4797–806.

    Article  CAS  Google Scholar 

  6. Fuenzalida MJ, Ruegg PL. Negatively controlled, randomized clinical trial to evaluate intramammary treatment of nonsevere, gram-negative clinical mastitis. J Dairy Sci. 2019;102:5438–57.

    Article  CAS  Google Scholar 

  7. Xiu L, Fu YB, Deng Y, Shi XJ, Bian ZY, Ruhan A, Wang X. Deep sequencing-based analysis of gene expression in bovine mammary epithelial cells after Staphylococcus aureus, Escherichia coli, and Klebsiella pneumoniae infection. Genet Mol Res. 2015;14:16948–65.

    Article  CAS  Google Scholar 

  8. Liu G, Liu Y, Ali T, Ferreri M, Gao J, Chen W, Yin J, Su J, Fanning S, Han B. Molecular and phenotypic characterization of Aerococcus viridans associated with subclinical bovine mastitis. PLoS One. 2015;10:e0125001.

    Article  Google Scholar 

  9. Gao J, Li S, Zhang J, Zhou Y, Xu S, Barkema HW, Nobrega DB, Zhu C, Han B. Prevalence of potential virulence genes in Klebsiella spp. isolated from cows with clinical mastitis on large Chinese dairy farms. Foodborne Pathog Dis. 2019;16:856–63.

    Article  Google Scholar 

  10. Lan J, Dou X, Li J, Yang Y, Xue C, Wang C, Gao N, Shan A. L-arginine ameliorates lipopolysaccharide-induced intestinal inflammation through inhibiting the TLR4/NF-κB and MAPK pathways and stimulating β-defensins expression in vivo and in vitro. J Agric Food Chem. 2020;68:2648–63.

    Article  CAS  Google Scholar 

  11. Zhao C, Wang Y, Yuan X, Sun G, Shen B, Xu F, Fan G, Jin M, Li X, Liu G. Berberine inhibits lipopolysaccharide-induced expression of inflammatory cytokines by suppressing TLR4-mediated NF-κB and MAPK signaling pathways in rumen epithelial cells of Holstein calves. J Dairy Res. 2019;86:171–6.

    Article  Google Scholar 

  12. Nyati KK, Masuda K, Zaman MM, Dubey PK, Millrine D, Chalise JP, Higa M, Li S, Standley D, Saito K, Hanieh H, Kishimoto T. TLR4-induced NF-κB and MAPK signaling regulate the IL-6 mRNA stabilizing protein Arid5a. Nucleic Acids Res. 2017;45:2687–703.

    Article  CAS  Google Scholar 

  13. Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517:311–20.

    Article  CAS  Google Scholar 

  14. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109.

    Article  CAS  Google Scholar 

  15. Bannerman DD, Paape M, Hare WR, Hope JC. Characterization of the bovine innate immune response to intramammary infection with Klebsiella pneumoniae. J Dairy Sci. 2004;87:2420–32.

    Article  CAS  Google Scholar 

  16. Kieffer N, Poirel L, Nordmann P, Madec JY, Haenni M. Emergence of colistin resistance in Klebsiella pneumoniae from veterinary medicine. J Antimicrob Chemother. 2015;70:1265–7.

    CAS  PubMed  Google Scholar 

  17. Saidani M, Messadi L, Soudani A, Daaloul-Jedidi M, Châtre P, Ben Chehida F, Mamlouk A, Mahjoub W, Madec J, Haenni M. Epidemiology, antimicrobial resistance, and extended spectrum beta-lactamase-producing enterobacteriaceae in clinical bovine mastitis in Tunisia. Microb Drug Resist. 2018;24:1242–8.

    Article  CAS  Google Scholar 

  18. Díaz-Muñoz SL, Koskella B. Bacteria-phage interactions in natural environments. Adv Appl Microbiol. 2014;89:135–83.

    Article  Google Scholar 

  19. Melo LDR, Oliveira H, Pires DP, Dabrowska K, Azeredo J. Phage therapy efficacy: a review of the last 10 years of preclinical studies. Crit Rev Microbiol. 2020;46:78–99.

    Article  CAS  Google Scholar 

  20. Martín-Díaz J, Lucena F, Blanch AR, Jofre J. Review. Indicator bacteriophages in sludge, biosolids, sediments and soils. Environ Res. 2020;182:109133.

    Article  Google Scholar 

  21. Lerondelle C, Poutrel B. Bacteriophage treatment trials on staphylococcal udder infection in lactating cows. Ann Rech Vet. 1980;11:421–6.

    CAS  PubMed  Google Scholar 

  22. Gill JJ, Pacan JC, Carson ME, Leslie KE, Griffiths MW, Sabour PM. Efficacy and pharmacokinetics of bacteriophage therapy in treatment of subclinical Staphylococcus aureus mastitis in lactating dairy cattle. Antimicrob Agents Chemother. 2006;50:2912–8.

    Article  CAS  Google Scholar 

  23. Malachowa N, Kobayashi SD, Porter AR, Freedman B, Hanley PW, Lovaglio J, Saturday GA, Gardner DJ, Scott DP, Griffin A, Cordova K, Long D, Rosenke R, Sturdevant DE, Bruno D, Martens C, Kreiswirth BN, Deleo FR. Vaccine protection against multidrug-resistant Klebsiella pneumoniae in a nonhuman primate model of severe lower respiratory tract infection. mBio. 2019;10:e02994-19.

    Article  Google Scholar 

  24. Motta C, Salmeri M, Anfuso CD, Amodeo A, Scalia M, Toscano MA, Giurdanella G, Alberqhina M, Lupoet G. Klebsiella pneumoniae induces an inflammatory response in an in vitro model of blood-retinal barrier. Infect Immun. 2014;82:851–63.

    Article  CAS  Google Scholar 

  25. Vareille-Delarbre M, Miquel S, Garcin S, Bertran T, Balestrino D, Evrard B, Forestier C. Immunomodulatory effects of lactobacillus plantarum on inflammatory response induced by Klebsiella pneumoniae. Infect Immun. 2019;87:e00570-19.

    Article  Google Scholar 

  26. Fuenzalida MJ, Ruegg PL. Molecular epidemiology of nonsevere clinical mastitis caused by Klebsiella pneumoniae occurring in cows on 2 Wisconsin dairy farms. J Dairy Sci. 2020;103:3479–92.

    Article  CAS  Google Scholar 

  27. Confer AW, Ayalew S. The OmpA family of proteins: roles in bacterial pathogenesis and immunity. Vet Microbiol. 2013;163:207–22.

    Article  CAS  Google Scholar 

  28. Chen W, Liu Y, Zhang L, Gu X, Liu G, Shahid M, Gao J, Ali T, Han B. Nocardia cyriacigeogica from bovine mastitis induced in vitro apoptosis of bovine mammary epithelial cells via activation of mitochondrial-caspase pathway. Front Cell Infect Microbiol. 2017;7:194.

    Article  CAS  Google Scholar 

  29. Wei Z, Wang J, Wang Y, Wang C, Liu X, Han Z, Fu Y, Zhang N. Effects of neutrophil extracellular traps on bovine mammary epithelial cells in vitro. Front Immunol. 2019;10:1003.

    Article  CAS  Google Scholar 

  30. Ali T, Ur Rahman S, Zhang L, Shahid M, Zhang S, Liu G, Gao J, Han B. ESBL-producing Escherichia coli from cows suffering mastitis in China contain clinical class 1 integrons with CTX-M linked to ISCR1. Front Microbiol. 2016;7:1931.

    PubMed  PubMed Central  Google Scholar 

  31. Szafrański SP, Winkel A, Stiesch M. The use of bacteriophages to biocontrol oral biofilms. J Biotechnol. 2017;250:29–44.

    Article  Google Scholar 

  32. Sordillo LM. Symposium review: Oxylipids and the regulation of bovine mammary inflammatory responses. J Dairy Sci. 2018;101:5629–41.

    Article  CAS  Google Scholar 

  33. Alva-Murillo N, Ochoa-Zarzosa A, López-Meza JE. Sodium octanoate modulates the innate immune response of bovine mammary epithelial cells through the TLR2/P38/JNK/ERK1/2 pathway: Implications during Staphylococcus aureus internalization. Front Cell Infect Microbiol. 2017;7:78.

    Article  Google Scholar 

  34. Caldeira MO, Bruckmaier RM, Wellnitz O. Meloxicam affects the inflammatory responses of bovine mammary epithelial cells. J Dairy Sci. 2019;102:10277–90.

    Article  CAS  Google Scholar 

  35. Zhang H, Jiang H, Fan Y, Chen Z, Li M, Mao Y, Karrow NA, Loor JJ, Moore S, Yang Z. Transcriptomics and iTRAQ-proteomics analyses of bovine mammary tissue with Streptococcus agalactiae-induced mastitis. J Agric Food Chem. 2018;66:11188–96.

    Article  CAS  Google Scholar 

  36. Ma N, Chang G, Huang J, Wang Y, Gao Q, Cheng X, Liu J, Shen X. Cis-9, trans-11-conjugated linoleic acid exerts an anti-inflammatory effect in bovine mammary epithelial cells after Escherichia coli stimulation through NF-κB signaling pathway. J Agric Food Chem. 2019;67:193–200.

    Article  CAS  Google Scholar 

  37. Wachi S, Kanmani P, Tomosada Y, Kobayashi H, Yuri T, Egusa S, Shimazu T, Suda Y, Aso H, Sugawara M, Saito T, Mishima T, Villena J, Ketazawaet H. Lactobacillus delbrueckii TUA4408L and its extracellular polysaccharides attenuate enterotoxigenic Escherichia coli-induced inflammatory response in porcine intestinal epitheliocytes via Toll-like receptor-2 and 4. Mol Nutr Food Res. 2014;58:2080–93.

    Article  CAS  Google Scholar 

  38. Li X, Fang F, Gao Y, Tang G, Xu W, Wang Y, Kong R, Tuyihong A, Wang Z. ROS induced by killer red. targeting mitochondria (mtKR) enhances apoptosis caused by radiation via Cyt c/Caspase-3 pathway. Oxid Med Cell Longev. 2019;2019:4528616.

  39. Wang HW, Zhang Y, Tan PP, Jia LS, Chen Y, Zhou BH. Mitochondrial respiratory chain dysfunction mediated by ROS is a primary point of fluoride-induced damage in Hepa1-6 cells. Environ Pollut. 2019;255:113359.

    Article  CAS  Google Scholar 

  40. Yang Y, Yu Y, Wang J, Li Y, Li Y, Wei J, Zheng T, Jin M, Sun Z. Silica nanoparticles induced intrinsic apoptosis in neuroblastoma SH-SY5Y cells via CytC/Apaf-1 pathway. Environ Toxicol Pharmacol. 2017;52:161–9.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was financially supported by Beijing Municipal Natural Science Foundation (No. 6192013), the National Natural Science Foundation of China (No. 31772813 and 31572587), the National Key R&D Project (No. 2016YFD0501203), Hebei Key R&D Project (19226607D) and the High-end Foreign Experts Recruitment Program (No. GDT20171100013). The above “Funding Organizations” provided financial support, and they had no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

BH, JG and JPK conceived and designed the experiment. YS and WZ performed the research and wrote the manuscript, GL, PC and YL performed the correlation of the genes to inflammation and apoptosis and associated interpretation, TA, YS and WZ wrote the first draft of the manuscript, JG assisted in the result analysis and re-edited the manuscript. BH and JPK revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Bo Han or Jian Gao.

Ethics declarations

Ethics approval and consent to participate

The current work was carried out according to ethical guidelines of China Agricultural University (CAU), Beijing, and approved by China Agricultural University Animal Ethics Committee (protocol CAUAEC-2010–0603). There were no vulnerable populations involved and no endangered species used in the experiment.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

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

Shi, Y., Zhao, W., Liu, G. et al. Bacteriophages isolated from dairy farm mitigated Klebsiella pneumoniae-induced inflammation in bovine mammary epithelial cells cultured in vitro. BMC Vet Res 17, 37 (2021). https://doi.org/10.1186/s12917-020-02738-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12917-020-02738-0

Keywords