Skip to main content

Dietary Zn proteinate with moderate chelation strength alleviates heat stress-induced intestinal barrier function damage by promoting expression of tight junction proteins via the A20/NF-κB p65/MMP-2 pathway in the jejunum of broilers

Abstract

Background

The aim of this study was to determine whether and how Zn proteinate with moderate chelation strength (Zn-Prot M) can alleviate heat stress (HS)-induced intestinal barrier function damage of broilers. A completely randomized design was used for comparatively testing the effects of Zn proteinate on HS and non-HS broilers. Under high temperature (HT), a 1 (Control, HT-CON) + 2 (Zn source) × 2 (added Zn level) factorial arrangement of treatments was used. The 2 added Zn sources were Zn-Prot M and Zn sulfate (ZnS), and the 2 added Zn levels were 30 and 60 mg/kg. Under normal temperature (NT), a CON group (NT-CON) and pair-fed group (NT-PF) were included.

Results

The results showed that HS significantly reduced mRNA and protein expression levels of claudin-1, occludin, junctional adhesion molecule-A (JAMA), zonula occludens-1 (ZO-1) and zinc finger protein A20 (A20) in the jejunum, and HS also remarkably increased serum fluorescein isothiocyanate dextran (FITC-D), endotoxin and interleukin (IL)-1β contents, serum diamine oxidase (DAO) and matrix metalloproteinase (MMP)-2 activities, nuclear factor kappa-B (NF-κB) p65 mRNA expression level, and protein expression levels of NF-κB p65 and MMP-2 in the jejunum. However, dietary supplementation with Zn, especially organic Zn as Zn-Prot M at 60 mg/kg, significantly decreased serum FITC-D, endotoxin and IL-1β contents, serum DAO and MMP-2 activities, NF-κB p65 mRNA expression level, and protein expression levels of NF-κB p65 and MMP-2 in the jejunum of HS broilers, and notably promoted mRNA and protein expression levels of claudin-1, ZO-1 and A20.

Conclusions

Our results suggest that dietary Zn, especially 60 mg Zn/kg as Zn-Prot M, can alleviate HS-induced intestinal barrier function damage by promoting the expression of TJ proteins possibly via induction of A20-mediated suppression of the NF-κB p65/MMP-2 pathway in the jejunum of HS broilers.

Background

As the global climate warms, animals suffer from more severe heat stress (HS) in the summer. The HS is one of the primary stressors that causes protein breakdown and ultimately results in aging and mortality [1]. The gastrointestinal tract is considered as the main target of HS [2]. Broilers are more susceptible to HS because they do not have sweat glands [3,4,5]. Previous studies have shown that HS damages the intestinal integrity and barrier function of broilers [6, 7], reducing health and growth performance and causing great economic losses in the poultry industry. Damage to the barrier function of the intestines is a primary factor leading to reduced growth rates and increased susceptibility to diseases [8].

In animals, intestinal barrier function of broilers is closely correlated to the expression levels of tight junction (TJ) proteins [9, 10]. Alhotan et al. [11] found that HS damages intestinal barrier function of broilers by reducing the mRNA expression levels of claudin-1, claudin-4, occludin and zonula occludens-1 (ZO-1). Deng et al. [12] reported that HS injures the barrier function of the small intestine by decreasing duodenal mRNA expression of occludin in broilers. Zhang et al. [13] showed that HS damages the intestinal barrier function of broilers by reducing the protein expression levels of claudin-1 and occludin. The above studies indicate that HS causes intestinal barrier dysfunction in broilers by decreasing the expression of TJ proteins. Heat stress-induced decrease in the expression of TJ proteins is associated with NF-κB activation in broilers [14], which in turn may promote the expression of matrix metalloproteinases 2 and 9 (MMPs 2 and 9) genes [15]. The MMPs are a class of endopeptidases that is responsible for degrading the TJ proteins, and only MMP-2 and MMP-9 are associated with the intestinal TJ proteins degradations [16, 17]. Therefore, HS may contribute to injuring small intestine barrier function in broilers possibly by suppressing TJ protein levels via activation of the NF-κB/MMP signaling pathway.

Food digestion and nutrient absorption are located mainly in the intestines [18]. The HS-induced intestinal barrier function damage causes digestive and absorption disorders as well as other diseases [19, 20]. Feed nutrition is one of the important measures to alleviate HS. Zinc (Zn) is an essential trace element for animals and human beings [21, 22]. Dietary Zn deficiency leads to decreased growth performance and feed utilization, impaired immune response and delayed wound healing [23,24,25]. Therefore, the addition of Zn to broiler diets to prevent Zn deficiency is required. The Zn, especially organic Zn, has potential benefits for alleviating HS-induced intestinal barrier function impairment in mammals [26] and in broilers [27,28,29]. The molecular mechanistic actions of inorganic Zn, have been investigated in other species [10, 30, 31]. Zinc oxide maintained the intestinal barrier function of weaning piglets by promoting the expression of TJ proteins [30]. Li et al. [10] reported that Zn oxide nanoparticles relieved HS-induced down-regulation of claudin and ZO-1 expressions in bovine intestinal epithelial cells in vitro by inhibiting the NF-κB pathway. Yan et al. [31] found that the Zn finger protein A20 (A20) is involved in the regulation of NF-κB expression in rats. In addition, a series of studies with broilers in our laboratory has demonstrated that organic Zn as Zn proteinate with moderate chelation strength (Zn-Prot M) displays better Zn absorption and the highest Zn bioavailability compared to Zn sulfate and other organic Zn sources [32,33,34,35]. However, whether and how organic Zn-Prot M can alleviate HS-induced intestinal barrier function damage in broilers has not been reported.

The jejunum has been shown to be the most sensitive segment of the small intestine in broilers in response to Zn and HS [36,37,38]. We thus hypothesized that Zn, especially the organic form of Zn as Zn-Prot M, could alleviate the HS-induced intestinal barrier function damage in broilers by promoting the expression of TJ proteins via induction of A20-mediated suppression of NF-κB/MMP (A20/NF-κB/MMP) signaling pathway in the jejunum of broilers. The aim of the present study was to determine the effects of dietary added Zn source and Zn level on growth performance, intestinal barrier function, expression of jejunal TJ proteins, and A20/NF-κB/MMP signaling pathway genes of HS broilers.

Materials and methods

Animal ethics

The experiment was approved by the Animal Ethics Committee of Yangzhou University [approval number: SYXK (Su) 2021-0027].

Experiment design and treatments

A completely randomized design was employed in this experiment. Under high temperature (HT, 9:00–17:00: 34 ± 1 °C, 8 h/d; 17:00–9:00: 28 ± 1 °C), a 1 (Control, HT-CON) + 2 [Zn source, ZnSO4·7H2O (ZnS) and Zn-Prot M (17.09% Zn and Qf = 51.6 by analysis)] × 2 [added Zn level, 30 and 60 mg/kg] factorial arrangement of treatments was used. The two dietary added Zn levels were based on previous studies [27, 28]. Under normal temperature (NT, 23 ± 1 °C, 24 h/d), a CON group (NT-CON) and pair-fed (NT-PF) group (the feed intake restriction to the same as that for the HT-CON group) were set up. Thus, there were a total of 7 different treatments (NT-CON, NT-PF, HT-CON, HT-ZnS 30, HT-ZnS 60, HT-Zn-Prot M 30 and HT-Zn-Prot M 60). The experimental treatments are shown in Table 1.

Table 1 The arrangement of experimental treatments for 22–42-day-old broilers

Animals and diets

A total of 1,150 one-day-old Arbor Acres (AA) male broilers with an average body weight (41.3 ± 0.1 g) were purchased from Jiangsu Jinghai Poultry Group Co., Ltd., China. All chickens were fed ad libitum with the same corn-soybean meal complete diet formulated based on the NRC [39] and Feeding standard of chicken in China [40] during d 1–21 (Table 2). At d 22 of age, 784 chickens were weighed, selected, and randomly allotted to 1 of 7 treatments with 7 replicates and 16 birds per replicate. The 16 chickens were placed in two adjacent stainless steel cages equipped with stainless steel feeders and waterers with 8 chickens per cage (90 cm × 70 cm × 45 cm). The two adjacent cages were regarded as one replicate unit. The broilers were maintained on a 24-h constant light schedule and handled in accordance with the Arbor Acres Broiler Management Guide [41], and allowed ad libitum access to diets (except for the NT-PF group chickens) and to tap water containing no detectable Zn. The room temperature for the NT-CON and NT-PF groups was maintained at 23 ± 1 °C daily, whereas the room temperature for the HT-CON, HT-ZnS-30, HT-ZnS-60, HT-Zn-Prot M-30 and HT-Zn-Prot M-60 groups was set as 34 ± 1 °C, 8 h/d, and the temperature for the remaining time was set as 28 ± 1 °C. The relative humidity under either NT or HT was maintained at 55% ± 5%. As the broilers in the HT-CON group had lower feed intake than those in the NT-CON group, to eliminate the potential effect of the reduced feed intake in the HT-CON group, the broilers in the NT-PF group were pair-fed the same amount of feed as those in the HT-CON group consumed on the previous day. Dead birds were recorded daily, and body weight and feed intake were measured per replicate unit on d 28, 35 and 42 to calculate the average daily feed intake (ADFI), average daily gain (ADG), feed-to-gain ratio (F/G), and mortality during d 22 to 42.

Table 2 Composition and nutrient levels of the broiler complete diets (as-fed basis)

The corn-soybean meal complete diets were formulated to meet or exceed the requirements of broilers for all nutrients as recommended by the NRC [39] and Feeding standard of chicken in China [40] (Table 2). The supplemental levels of Zn as ZnS in the above basal complete diets as shown in Table 2, were based on the recent research results on dietary Zn requirements of broilers fed the conventional corn-soybean meal diets from 1 to 21 days of age or 22 to 42 days of age from our laboratory [42, 43]. During d 22–42, a single batch of the complete diet (CON) was mixed and then divided into 7 aliquots according to the experimental treatments. Either ZnS or Zn-Prot M was added to the above CON diet to formulate the respective treatment diets. The ZnS was reagent grade (Sinopharm Chemical Reagent Co. Ltd., Shanghai, China) and contained 22.49% Zn on a basis of analysis (purity > 99.5%). The Zn-Prot M was the same as that used in the study of Hu et al. [35], which was provided by a special commercial company and contained 17.09% Zn with the chelation strength (Qf value) of 51.6 on a basis of analysis. The Qf value of 51.6 is categorized as a moderate chelation strength based on the classification of Holwerda et al. [44]. Lysine and methionine levels in the control diet or the diet supplemented with the ZnS were balanced by adding synthetic lysine-HCl and DL-methionine based on supplemental amounts of lysine and methionine from the Zn-Prot M. The analyzed Zn contents in all treatment diets are shown in Table 1.

Sample collection and preparation

Diet samples were collected and analyzed for calcium, Zn, or crude protein contents. The tap water was collected for analysis of Zn content. At 28, 35 and 42 days of age, 3 chickens from each replicate unit were selected based on the average body weight of the cage unit and then anesthetized by intraperitoneal injection of propofol (20 mg/kg body weight). As the biomarkers from the hepatic portal vein blood better reflect the intestinal barrier function and health status of animals, blood samples were collected from the hepatic portal vein of each bird, and serum was separated and stored at –20 °C for analyses of endotoxin, IL-1β and IL-6 contents and diamine oxidase (DAO) activity. The chickens were euthanized by decapitation after taking blood, and the jejunum was separated and rinsed with ice-cold saline solution. The jejunal mucosa were scraped with a sterilized microscope slide and then placed in sterilized 1.5-mL centrifuge tubes, immediately frozen in liquid nitrogen and stored at –80 °C for analyses of mRNA and protein expression levels. In addition, another chicken was selected from each replicate unit based on the average body weight of the cage unit, and subjected to fluorescein isothiocyanate dextran (FITC-D, catalog number FD4; Merck, Darmstadt, DEU) oral perfusion in a dose of 8.32 mg/kg body mass [45]. At 1 h after perfusion, blood samples were collected from the hepatic portal vein, and centrifuged at 3,000 r/min and 4 °C to obtain serum samples for analysis of FITC-D content. All samples from 3 birds in each replicate unit were pooled into 1 sample before analyses.

Analyses of dietary crude protein, calcium and Zn contents, serum indices and jejunal MMP-2 activity

The crude protein content in the diets was determined using the Kjeldahl method [46]. The 5110 inductively coupled plasma optical emission spectrometry (Agilent Technologies, Mulgrave, Australia) was used to measure the calcium content in the diets, and Zn content in the diets and tap water as described previously [47]. Commercial kits were used to detect serum DAO activity (catalog number BC1285; Solarbio, Beijing, China), endotoxin (catalog number EC80545S; Bioendo, Xiamen, China), IL-1β (catalog number SEKCN-0153; Solarbio, Beijing, China) and IL-6 (catalog number SEKCN-0161; Solarbio, Beijing, China) contents, and jejunal MMP-2 activity (catalog number SEKCN-0022; Solarbio, Beijing, China). Serum FITC-D was measured by a multifunctional microplate reader as previously described [45].

Quantitative real-time PCR

Total RNA was extracted from 50 mg jejunum using the TRIzol kit (catalog number 15596018CN; Invitrogen, Carlsbad, California, USA). A reverse transcription kit (catalog number R223-01; Vazyme, Nanjing, China) was used to reverse transcribe the RNA into cDNA, and quantitative real-time PCR was performed using SYBR Green (catalog number Q111-02; Vazyme, Nanjing, China). All primers (Table 3) were synthesized by Tsingke Biotech (Beijing, China). β-Actin and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were used as reference genes. The data were analyzed using the 2−ΔΔCt relative expression method [48].

Table 3 Primer sequences for real-time PCR amplification

Total protein extraction and Western blotting

Frozen jejunal mucosa samples were homogenized in ice-cold RIPA lysis buffer (catalog number R0010; Solarbio, Beijing, China) as previously described [49]. The protein concentration was determined by BCA protein assessment kit (catalog number 23225; ThermoFisher, Waltham, Massachusetts, USA). Proteins were separated by SDS-PAGE, and transferred onto nitrocellulose (NC) membranes. The NC membrane was blocked at room temperature for 1.5 h using 5% BSA. After that, it was incubated overnight at 4 °C with primary antibodies [claudin-1 (catalog number WL03073; Wanlei, Shenyang, China; 1:1,000), occludin (catalog number 331500; Invitrogen, Carlsbad, California, USA; 1:1,000), ZO-1 (catalog number 617300; Life technologies, Carlsbad, California, USA; 1:250), JAMA (catalog number A1241; ABclonal, Wuhan, China; 1:1,000), A20 (catalog number WL00820; Wanlei, Shenyang, China; 1:500), NF-κB p65 (catalog number 10745-1-AP; Proteintech, Wuhan, China; 1:3,000), MMP-2 (catalog number bs-0412R; Bioss, Beijing, China; 1:1,000), MMP-9 (catalog number bs-4593R; Bioss, Beijing, China; 1:1,000), Lamin A/C (catalog number BS1446; Bioworld, Nanjing, China; 1:1,000) and β-actin (catalog number HX1827; Huaxingbio, Beijing, China; 1:5,000)]. On the following day, Tris-Buffered Saline and Tween 20 (TBST) were used to wash the membranes and then they were incubated with secondary antibody. The protein bands were visualized using the chemiluminescence (ECL) detection kit (catalog number 180-5001; Tanon, Beijing, China). Quantification of protein bands was performed with Tanon GIS 1D software (Tanon, Beijing, China). Protein quantification was performed using β-actin as the cytoplasmic internal reference and Lamin A/C as the nuclear internal reference.

Statistical analyses

All data were analyzed statistically using SAS software (version 9.4, 2013). The data obtained from NT-CON, NT-PF and HT-CON groups were analyzed using the GLM procedure through one-way ANOVA. A single degree of freedom contrast was used to test the differences between all supplemental Zn treatments and the HT-CON. The data excluding the HT-CON under HT were analyzed by two-way ANOVA using the GLM procedure, and the statistical model included Zn source, added Zn level and their interaction. Differences among means were tested by the least significant difference (LSD) method. The replicate cage unit was the experimental unit and the statistical significance was set at P < 0.05.

Results

Growth performance and mortality

As shown in Fig. 1, the treatment groups NT-CON, NT-PF and HT-CON did not affect mortality, but influenced (P < 0.01) ADG, ADFI and F/G of broilers during d 22–42. Compared with the NT-CON, HT-CON and NT-PF groups exhibited decreased (P < 0. 05) ADG and ADFI, but increased (P < 0.05) F/G with no differences between HT-CON and NT-PF groups. Compared with the HT-CON, all supplemental Zn treatments under HT had no effect on the growth performance indices (ADG, ADFI and F/G) and mortality of broilers from 22 to 42 days of age under HT. The Zn source, added Zn level and their interaction had no effects on the growth performance indices and mortality of broilers from 22 to 42 days of age under HT.

Fig. 1
figure 1

Effects of dietary added Zn source and level on growth performance and mortality of HS broilers from 22 to 42 days of age. A ADFI. B ADG. C F/G. D Mortality. ZnS: Zn sulfate. Zn-Prot M: Zn proteinate with moderate chelation strength (Qf  = 51.6). NT: normal temperature, 23 ± 1 °C, 24 h/d. HT: high temperature, 9:00–17:00: 34 ± 1 °C, 8 h/d, and 17:00–9:00: 28 ± 1 °C. ADFI: average daily feed intake. ADG: average daily gain. F/G: feed to gain ratio. CON: control group. PF: pair-fed group, and the feed intake of PF birds was restricted to match that of those in the HT-CON group. Different letters indicate significant differences among treatments (P < 0.05). Data are mean ± SE (n = 5–7)

FITC-D, endotoxin and interleukin contents and DAO activity in serum, and jejunal MMP-2 activity

As shown in Fig. 2, the treatment groups NT-CON, NT-PF and HT-CON did not affect serum DAO activity of broilers on d 28, but influenced (P < 0.04) serum FITC-D contents and DAO activities on d 35 and 42, and serum FITC-D content on d 28, and serum endotoxin content on d 42. Compared with the NT-CON, HT-CON and NT-PF increased (P < 0.05) serum FITC-D contents of broilers on d 28 and 42, and serum DAO activity on d 42. Compared with the NT-PF, HT-CON increased (P < 0.05) serum FITC-D content and DAO activities of broilers on d 35 and 42, and serum endotoxin content on d 42. Compared with the HT-CON, all supplemental Zn treatments under HT decreased (P < 0.05) serum DAO activities of broilers on d 28, 35 and 42, and serum FITC-D and endotoxin contents of broilers on d 42. Under HT, Zn source affected (P < 0.05) serum endotoxin content of broilers on d 42. Added Zn level affected (P < 0.05) serum FITC-D and endotoxin contents and DAO activity of broilers on d 42, and serum FITC-D content on d 35. There were no interactions between Zn source and added Zn level in all of the above indices. Compared with the ZnS, Zn-Prot M decreased (P < 0.05) serum endotoxin content in broilers on d 42. Compared with the 30 mg/kg of supplemental Zn, 60 mg/kg decreased (P < 0.05) serum FITC-D and endotoxin contents and DAO activity of broilers on d 42, and serum FITC-D content on d 35.

Fig. 2
figure 2

Effects of dietary added Zn source and level on serum FITC-D contents and DAO activities of HS broilers on d 28, 35 and 42 and serum endotoxin content of HS broilers on d 42. A Serum FITC-D content of broilers on d 28. B Serum DAO activity of broilers on d 28. C Serum FITC-D content of broilers on d 35. D Serum DAO activity of broilers on d 35. E Serum FITC-D content of broilers on d 42. F Serum DAO activity of broilers on d 42. G Serum endotoxin content of broilers on d 42. ZnS: Zn sulfate. Zn-Prot M: Zn proteinate with moderate chelation strength (Qf  = 51.6). NT: normal temperature, 23 ± 1 °C, 24 h/d. HT: high temperature, 9:00–17:00: 34 ± 1 °C, 8 h/d, and 17:00–9:00: 28 ± 1 °C. CON: control group. PF: pair-fed group, and the feed intake of PF birds was restricted to match that of those in the HT-CON group. FITC-D: fluorescein isothiocyanate dextran. DAO: diamine oxidase. Different letters indicate significant differences among treatments (P < 0.05). *P < 0.05. Data are mean ± SE (n = 4–7)

As shown in Fig. 3, the treatment groups NT-CON, NT-PF and HT-CON had no effect on serum IL-6 content, but influenced (P < 0.05) serum IL-1β content and jejunal MMP-2 activity of broilers on d 42. Compared with the NT-CON, HT-CON increased (P < 0.05) serum IL-1β content and jejunal MMP-2 activity. Compared with the NT-PF, HT-CON increased (P < 0.05) serum IL-1β, but did not change jejunal MMP-2 activity. Compared with the HT-CON, all supplemental Zn treatments under HT decreased (P < 0.01) jejunal MMP-2 activity, and had no effect on serum IL-1β and IL-6 contents. Under HT, Zn source did not affect serum IL-6 content, but affected (P < 0.05) serum IL-1β content and jejunal MMP-2 activity. Added Zn level and its interaction with Zn source did not affect serum IL-1β and IL-6 contents, and jejunal MMP-2 activity. Compared with the ZnS, Zn-Prot M decreased (P < 0.05) serum IL-1β content and jejunal MMP-2 activity.

Fig. 3
figure 3

Effects of dietary added Zn source and level on serum IL-1β and IL-6 contents and jejunal MMP-2 activity of HS broilers on d 42. A Serum IL-1β content. B Serum IL-6 content. C Jejunal MMP-2 activity. ZnS: Zn sulfate. Zn-Prot M: Zn proteinate with moderate chelation strength (Qf = 51.6). NT: normal temperature, 23 ± 1 °C, 24 h/d. HT: high temperature, 9:00–17:00: 34 ± 1 °C, 8 h/d, and 17:00–9:00: 28 ± 1 °C. CON: control group. PF: pair-fed group, and the feed intake of PF birds was restricted to match that of those in the HT-CON group. IL, interleukin. MMP, matrix metalloproteinase. Different letters indicate significant differences among treatments (P < 0.05). Data are mean ± SE (n = 5–7)

mRNA expression levels

As shown in Fig. 4, the treatment groups NT-CON, NT-PF and HT-CON had no effect on mRNA expression levels of MMP-2 and MMP-9, but influenced (P < 0.01) mRNA expression levels of claudin-1, occludin, JAMA, ZO-1, A20 and NF-κB p65 in the jejunum of broilers on d 42. Compared with the NT-CON, either NT-PF or HT-CON decreased (P < 0.01) mRNA expression levels of JAMA and A20, but increased (P < 0.0001) NF-κB p65 mRNA expression. Compared with the NT-PF, HT-CON decreased (P < 0.05) mRNA expression levels of claudin-1, occludin, JAMA and ZO-1, and increased (P < 0.05) NF-κB p65 mRNA expression. Compared with the HT-CON, all supplemental Zn treatments under HT increased (P < 0.05) claudin-1 and A20 mRNA expression levels, and decreased (P < 0.0001) NF-κB p65 mRNA expression. Under HT, Zn source did not affect mRNA expression levels of claudin-1, occludin, JAMA, ZO-1, MMP-2 and MMP-9, but influenced (P < 0.05) mRNA expression levels of A20 and NF-κB p65. Added Zn level did not affect mRNA expression levels of occludin, JAMA, MMP-2 and MMP-9, but influenced (P < 0.05) mRNA expression levels of claudin-1, ZO-1, A20 and NF-κB p65. No interactions between Zn source and added Zn level were detected on mRNA expression levels for any of the above-mentioned genes. Compared with the ZnS, Zn-Prot M increased (P < 0.05) A20 mRNA expression and decreased (P < 0.05) NF-κB p65 mRNA expression. Compared with the 30 mg/kg of supplemental Zn, 60 mg/kg increased (P < 0.05) mRNA expression levels of claudin-1, ZO-1 and A20, and decreased (P < 0.01) NF-κB p65 mRNA expression.

Fig. 4
figure 4

Effects of dietary added Zn source and level on mRNA expression levels of tight junction proteins and target proteins in the related signaling pathway in the jejunum of HS broilers at 42 days of age. A–D mRNA expression levels of related tight junction proteins (claudin-1, occludin, JAMA and ZO-1). E–H mRNA expression levels of target proteins (A20, NF-ΚB p65, MMP-2 and MMP-9) in A20/NF-κB p65/MMP-2 signaling pathway. ZnS: Zn sulfate. Zn-Prot M: Zn proteinate with moderate chelation strength (Qf = 51.6). NT: normal temperature, 23 ± 1 °C, 24 h/d. HT: high temperature, 9:00–17:00: 34 ± 1 °C, 8 h/d, and 17:00–9:00: 28 ± 1 °C. CON: control group. PF: pair-fed group, and the feed intake of PF birds was restricted to match that of those in the HT-CON group. ZO-1, zonula occludens-1; JAMA, junctional adhesion molecule-A; MMP, matrix metalloproteinase; A20, Zn finger protein A20; NF-κB, nuclear factor kappa-B. Values of mRNA abundance levels of target genes were calculated as the relative quantities (RQ) of claudin-1, occludin, JAM-A, ZO-1, MMP2, MMP9, A20 or NF-κB p65 mRNA to the geometric mean of internal reference genes β-actin and GAPDH mRNA using 2Ct. Different letters indicate significant differences among treatments (P < 0.05). *P < 0.05. Data are mean ± SE (n = 5–7)

Protein expression levels

As shown in Fig. 5, the treatment groups NT-CON, NT-PF and HT-CON had no effect on MMP-9 protein expression, but influenced (P < 0.0001) protein expression levels of claudin-1, occludin, JAMA, ZO-1, A20, NF-κB p65 and MMP-2 in the jejunum of broilers on d 42. Compared with the NT-CON, both NT-PF and HT-CON decreased (P < 0.05) protein expression levels of claudin-1, occludin, JAMA, ZO-1 and A20, but increased MMP-2 protein expression. Compared with the NT-PT, HT-CON decreased (P < 0.05) claudin-1, occludin, JAMA, ZO-1 and A20, and increased NF-κB p65 protein expression. Compared with the HT-CON, all supplemental Zn treatments under HT increased (P < 0.01) protein expression levels of claudin-1, occludin, JAMA, ZO-1 and A20, but decreased (P < 0.0001) protein expression levels of NF-κB p65 and MMP-2. Under HT, Zn source did not affect protein expression levels of ZO-1, A20, NF-κB p65 and MMP-9, but influenced (P < 0.05) protein expression levels of claudin-1, occludin, JAMA and MMP-2. Added Zn level did not affect protein expression levels of ZO-1 and MMP-9, but influenced (P < 0.05) protein expression levels of claudin-1, occludin, JAMA, A20, NF-κB p65 and MMP-2. No interactions between Zn source and added Zn level were detected on protein expression levels for any of the above-mentioned genes. Compared with the ZnS, Zn-Prot M increased (P < 0.05) protein expression levels of claudin-1, occludin and JAMA, but decreased (P < 0.05) MMP-2 protein expression. Compared with the 30 mg/kg of supplemental Zn, 60 mg/kg increased (P < 0.05) protein expression levels of claudin-1, occludin, JAMA and A20, and decreased (P < 0.0001) protein expression levels of NF-κB p65 and MMP-2.

Fig. 5
figure 5

Effects of dietary added Zn source and level on protein expression levels of tight junction proteins and target proteins in the related signaling pathway in the jejunum of HS broilers at 42 days of age. A–D Protein expression levels of related tight junction proteins (claudin-1, occludin, JAMA and ZO-1). E–H Protein expression levels of target proteins (A20, NF-ΚB p65, MMP-2 and MMP-9) in A20/NF-κB p65/MMP-2 signaling pathway. I Representative immunoblots of related tight junction proteins (claudin-1, occludin, JAMA and ZO-1). J Representative immunoblots of target proteins (A20, NF-ΚB p65, MMP-2 and MMP-9) in A20/NF-κB p65/MMP-2 signaling pathway. ZnS: Zn sulfate. Zn-Prot M: Zn proteinate with moderate chelation strength (Qf  = 51.6). NT: normal temperature, 23 ± 1℃, 24 h/d. HT: high temperature, 9:00–17:00: 34 ± 1 °C, 8 h/d, and 17:00–9:00: 28 ± 1 °C. CON: control group. PF: pair-fed group, and the feed intake of PF birds was restricted to match that of those in the HT-CON group. ZO-1, zonula occludens-1; JAMA, junctional adhesion molecule-A. MMP, matrix metalloproteinase; A20, Zn finger protein A20; NF-κB, nuclear factor kappa-B. Values of target protein expression levels were expressed by the relative quantities (RQ) of target protein band intensity to the internal reference band intensity of β-actin or Lamin A/C. Different letters indicate significant differences among treatments (P < 0.05). *P < 0.05. Data are mean ± SE (n = 5–7)

Discussion

The results from the current study show that HS significantly reduced ADFI, ADG, mRNA expression levels of claudin-1, occludin, JAMA, ZO-1 and A20, and their corresponding protein expression, and remarkably increased F/G, serum FITC-D, endotoxin and IL-1β contents, serum DAO activity as well as MMP-2 activity, NF-κB p65 mRNA expression level, and protein expression levels of NF-κB p65 and MMP-2 in the jejunum of broilers. However, dietary supplementation of Zn, especially organic Zn as Zn-Prot M at 60 mg/kg, significantly decreased serum FITC-D, endotoxin and IL-1β contents, serum DAO activity as well as MMP-2 activity, NF-κB p65 mRNA expression level, and protein expression levels of NF-κB p65 and MMP-2, and notably promoted mRNA expression levels of claudin-1, ZO-1 and A20, and protein expression levels of claudin-1, occludin, JAMA, ZO-1 and A20 in the jejunum of HS broilers. The above results suggest that dietary supplemental Zn, especially 60 mg Zn/kg as Zn-Prot M, could alleviate the HS-induced intestinal barrier function damage by promoting the expression of TJ proteins via induction of A20-mediated suppression of the NF-κB p65/MMP-2 pathway (A20/NF-κB p65/MMP-2 pathway) in the jejunum of HS broilers, which supports our hypothesis. These findings have not been reported before, and thus provide a scientific foundation for the alleviation of the HS-induced intestinal barrier function injury through dietary optimal Zn addition in broiler production.

Previous study has indicated that HS has a negative impact on the growth performances of broilers [50]. In the present study, HS decreased ADG and ADFI, and increased F/G of broilers during d 22–42 mainly due to a reduced feed intake, which is consistent with previous findings [51, 52]. Some of the previous studies showed that dietary Zn addition can alleviate HS-induced growth retardation [53, 54]. In other studies, dietary Zn supplementation had no significant effect on the growth performance of broilers exposed to HS [55, 56], which was consistent with our present results. It appears that the effect of dietary Zn on the growth performance of heat-stressed broilers is dependent on the Zn level in basal diets, the dose of supplemental Zn, dietary formulation, and experimental duration time [57, 58].

Earlier studies have demonstrated that HS can impair intestinal barrier function in broilers [59, 60], and that dietary Zn could improve intestinal barrier function [29, 61]. FITC-D, a macromolecular substance, cannot cross the intestinal epithelial barrier under normal conditions [62]. Hence, oral administration of FITC-D has been used as an indicator of intestinal permeability in broilers [63]. Furthermore, when intestinal barrier function is compromised, DAO and endotoxin can be released into blood. Therefore, serum DAO activity and endotoxin content are also regarded as typical indicators for reflecting the intestinal barrier function [64,65,66]. Alhenaky et al. [59], Tabler et al. [45] and Lan et al. [67] reported that HS-induced damage of intestinal barrier function in broilers is accompanied by an increase in endotoxin and FITC-D contents and DAO activity in serum. Similar results were obtained in the present study. The HS-induced intestinal barrier function damage was caused by the combined effect of both the reduced feed intake and HT itself, but the effect of HT itself was greater than that of the reduced feed intake, which is in line with one previous report [60]. Dietary Zn was shown to alleviate the HS-induced increases in endotoxin content in pig serum [68], FITC-D content in broiler serum [69] and DAO activity in duck serum [70]. Similar results were observed in our study in which dietary Zn supplementation was observed to relieve HS-induced increases in serum FITC-D and endotoxin contents and DAO activity. Furthermore, Zn-Prot M was more effective than ZnS in decreasing the serum endotoxin content of HS broilers, while 60 mg/kg of added Zn was more effective than 30 mg/kg of added Zn in decreasing serum FITC-D and endotoxin contents and DAO activity of broilers, which has not been reported before. Our findings, together with those of others [28, 71, 72], indicate that dietary supplemental Zn, especially 60 mg Zn/kg as Zn-Prot M, can protect the intestinal barrier of broilers from HS-induced damage.

The TJ proteins are pivotal structures for maintaining intestinal integrity and barrier function [73]. Ruff et al. [74] reported that the damage of the small intestine is aggravated with increased HS exposure time. Siddiqui et al. [75] demonstrated that the damage of HS on the jejunum of broilers was more obvious. Therefore, the jejunum of broilers at 42 days of age was selected as the target small intestinal segment in the subsequent measurements of the present study. Alhotan et al. [11] reported that HS damages intestinal barrier function by reducing the expression of claudin-1, occludin and JAMA in the jejunum of broilers. Xia et al. [76] demonstrated that the HS-induced damage of intestinal barrier function is accompanied by decreased mRNA expression levels of claudin-1 and ZO-1 in pigs. In the present study, we found that HS injures intestinal barrier function partially by down-regulating the expression of claudin-1, occludin, JAMA, and ZO-1 in the jejunum of broilers. Additionally, HS-induced down-regulation in the expression of the above TJ proteins in the jejunum of broilers was attributed to the combined effect of both the reduced feed intake and HT itself, but the effect of HT itself was greater than that of the reduced feed intake. In a study by Xie et al. [77], ZnS supplementation was found to attenuate lipopolysaccharide-induced decreases of claudin-1, occludin and ZO-1 mRNA and protein expression in duck jejunal epithelial cells. Similar results were observed in the current study in which dietary Zn addition relieved HS-induced down-regulation of claudin-1, occludin, JAM-A and ZO-1 protein expression levels in the jejunum of broilers. Moreover, the Zn-Prot M was more effective than ZnS, and 60 mg/kg of supplemental Zn was better than 30 mg/kg. The above results indicate that dietary supplemental Zn, especially 60 mg Zn/kg as Zn-Prot M, can alleviate HS-induced down-regulation of main TJ protein expression in the jejunum of broilers, which has not been reported before. However, the mechanism by which Zn regulates the expression of TJ proteins needs further investigation.

It is reported that the decreased protein expression of TJ proteins is closely correlated with the activation of NF-κB [78]. The NF-κB signaling pathway plays a crucial role in inflammation [79]. The activation of NF-kB promotes the production of inflammatory cytokines TNFα, IL-1β, and IL-6 [80]. In the present study, we found that HS increased the IL-1β content in the serum of broilers mainly due to the effect of HT itself, reflecting NF‐kB activation. A study with bovines indicated that HS-induced decrease in TJ protein expression is associated with NF-κB activation [10]. Furthermore, previous studies revealed that NF-κB is a key transcriptional factor participating in the regulation of MMP-2 and MMP-9 gene expression [81, 82]. Both MMP-2 and MMP-9 are responsible for degrading the intestinal TJ proteins [16, 17]. In the present study, HS increased the jejunal NF-κB p65 and MMP-2 protein expression levels and also MMP-2 activity mainly due to the effect of HT itself. Dietary Zn addition alleviated the HS-induced down-regulation of the jejunal NF-κB p65 mRNA and protein expression levels as well as the level of MMP-2 protein expression and MMP-2 activity. Additionally, the remission effect of Zn-Prot M was greater than that of ZnS, and the remission effect of supplemental Zn was greater at 60 mg/kg than at 30 mg/kg. The above findings suggest that dietary supplemental Zn, especially 60 mg Zn/kg as Zn-Prot M, can alleviate HS-induced down-regulation of main TJ protein expression possibly by inhibiting the NF-κB p65/MMP-2 signaling pathway in the jejunum of broilers, which has not been reported before.

A20 mediates some of the biological actions of Zn [83], and it is required for the Zn-induced NF-κB inactivation [84]. Li et al. [85] reported that dietary Zn supplementation inhibits intestinal inflammation via A20-mediated NF-kB inactivation. In the current investigation, we found that HS markedly decreased A20 mRNA and its protein expression in the jejunum of broilers mainly due to the effect of HT itself, but dietary Zn addition alleviated the HS-induced down-regulation of the jejunal A20 mRNA and its protein expression. Moreover, the remission effect of Zn-Prot M was greater than that of ZnS in A20 mRNA expression, and the remission effect of supplemental Zn was greater at 60 mg/kg than at 30 mg/kg in A20 mRNA and its protein expressions. Therefore, dietary supplemental Zn, especially 60 mg Zn/kg as Zn-Prot M, can alleviate HS-induced down-regulation of A20 expression in the jejunum of broilers and thereby allow restoration of A20-mediated suppression of the NF-κB p65/MMP-2 pathway (A20/NF-κB p65/MMP-2 pathway), which has not been reported before.

Conclusions

Dietary supplementation with Zn, especially organic Zn as Zn-Prot M at 60 mg/kg, alleviates HS-induced intestinal barrier function damage by promoting the expression of TJ proteins possibly via induction of A20-mediated suppression of NF-κB p65/MMP-2 pathway (A20/NF-κB p65/MMP-2 pathway) in the jejunum of broilers. Further studies need to be conducted using a primary cultured jejunal epithelial cell model or the jejunal organoid model of broilers as well as gene over-expression and RNA interference to address and confirm the above possible mechanisms.

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

AA:

Arbor Acres

ADFI:

Average daily feed intake

ADG:

Average daily gain

A20:

Zinc finger protein A20

DAO:

Diamine oxidase

FITC-D:

Fluorescein isothiocyanate dextran

F/G:

Feed to gain ratio   

HS:

Heat stress

IL:

Interleukin

JAMA:

Junctional adhesion molecule A

MMP:

Matrix metalloproteinase

NF-κB p65:

Nuclear factor kappa-B p65

TJ:

Tight junction protein

ZO-1:

Zonula occludens-1

References

  1. Schaller A. A cut above the rest: The regulatory function of plant proteases. Planta. 2004;220:183–97. https://doi.org/10.1007/s00425-004-1407-2.

    Article  CAS  PubMed  Google Scholar 

  2. Wasti S, Sah N, Singh AK, Lee CN, Jha R, Mishra B. Dietary supplementation of dried plum: a novel strategy to mitigate heat stress in broiler chickens. J Anim Sci Biotechnol. 2021;12:58. https://doi.org/10.1186/s40104-021-00571-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sahin K, Sahin N, Kucuk O, Hayirli A, Prasad AS. Role of dietary zinc in heat-stressed poultry: a review. Poult Sci. 2009;88:2176–83. https://doi.org/10.3382/ps.2008-00560.

    Article  CAS  PubMed  Google Scholar 

  4. Wasti S, Sah N, Mishra B. Impact of heat stress on poultry health and performances, and potential mitigation strategies. Animals. 2020;10:1266–85. https://doi.org/10.3390/ani10081266.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Wang HS, Ni XQ, Qing XD, Liu L, Xin JG, Luo M, et al. Probiotic lactobacillus johnsonii BS15 improves blood parameters related to immunity in broilers experimentally infected with subclinical necrotic enteritis. Front Microbiol. 2018;9:49. https://doi.org/10.3389/fmicb.2018.00049.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Peng XY, Xing T, Li JL, Zhang L, Jiang Y, Gao F. Guanidinoacetic acid supplementation improves intestinal morphology, mucosal barrier function of broilers subjected to chronic heat stress. J Anim Sci. 2023;101:skac355 https://doi.org/10.1093/jas/skac355.

    Article  PubMed  Google Scholar 

  7. Du MF, Cheng YF, Chen YP, Wang SQ, Zhao HR, Wen C, et al. Dietary supplementation with synbiotics improves growth performance, antioxidant status, immune function, and intestinal barrier function in broilers subjected to cyclic heat stress. Environ Sci Pollut Res. 2023;30:18026–38. https://doi.org/10.1007/s11356-022-23385-y.

    Article  CAS  Google Scholar 

  8. Liu KH, Liu ES, Lin L, Hu Y, Yuan Y, Xiao WY. L-theanine mediates the p38MAPK signaling pathway to alleviate heat-induced oxidative stress and inflammation in mice. Food Funct. 2022;13:2120–30. https://doi.org/10.1039/d1fo03077a.

    Article  CAS  PubMed  Google Scholar 

  9. Proszkowiec WM, Schreier LL, Kahl S, Miska KB, Russell B, Elsasser TH. Effect of delayed feeding post-hatch on expression of tight junction and gut barrier-related genes in the small intestine of broiler chickens during neonatal development. Poult Sci. 2020;99:4714–29. https://doi.org/10.1016/j.psj.2020.06.023.

    Article  CAS  Google Scholar 

  10. Li YX, Pan MY, Meng SD, Xu WH, Wang SC, Dou MY, et al. The effects of zinc oxide nanoparticles on antioxidation, inflammation, tight junction integrity, and apoptosis in heat-stressed bovine intestinal epithelial cells in vitro. Biol Trace Elem Res. 2023;202:2042–51. https://doi.org/10.1007/s12011-023-03826-6.

    Article  CAS  PubMed  Google Scholar 

  11. Alhotan RA, Sulaiman ARA, Alharthi AS, Abudabos AM. Protective influence of betaine on intestinal health by regulating inflammation and improving barrier function in broilers under heat stress. Poult Sci. 2021;100:101337. https://doi.org/10.1016/j.psj.2021.101337.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Deng CX, Zheng J, Zhou H, You JM, Li GH. Dietary glycine supplementation prevents heat stress-induced impairment of antioxidant status and intestinal barrier function in broilers. Poult Sci. 2023;102:102408. https://doi.org/10.1016/j.psj.2022.102408.

    Article  CAS  PubMed  Google Scholar 

  13. Zhang C, Zhao XH, Yang L, Chen XY, Jiang RS, Jin SH, et al. Resveratrol alleviates heat stress-induced impairment of intestinal morphology, microflora, and barrier integrity in broilers. Poult Sci. 2017;96:4325–32. https://doi.org/10.3382/ps/pex266.

    Article  CAS  PubMed  Google Scholar 

  14. Wang Z, Zhang DW, Yu WJ, Qi CH, Yuan J. Probiotic cocktails alleviate heat-induced intestinal barrier dysfunction and oxidative stress in broiler chickens. Anim Husbandry Feed Sci. 2019;11:107–14 https://doi.org/10.19578/j.cnki.ahfs.2019.03-04.005.

    Google Scholar 

  15. Li J, Lau GK, Chen LL, Dong SS, Lan HY, Huang XR, et al. Interleukin 17A promotes hepatocellular carcinoma metastasis via NF-κB induced matrix metalloproteinases 2 and 9 expression. PLoS ONE. 2011;6:e21816. https://doi.org/10.1371/journal.pone.0021816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Walshe M, Nayeri S, Ji JY, Hernandez-Rocha C, Sabic K, Hu LY, et al. A role for CXCR3 ligands as biomarkers of post-operative crohn’s disease recurrence. J Crohns & Colitis. 2022;16:900–10. https://doi.org/10.1093/ecco-jcc/jjab186.

    Article  Google Scholar 

  17. McKittrick CM, Lawrence CE, Carswell HV. Mast cells promote blood brain barrier breakdown and neutrophil infiltration in a mouse model of focal cerebral ischemia. J Cereb Blood Flow Metab. 2015;35:638–47. https://doi.org/10.1038/jcbfm.2014.239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tao S, Li L, Ding JN, Zhong JJ, Lu Y, Wang XL, et al. Mobilization of soil-bound residue of organochlorine pesticides and polycyclic aromatic hydrocarbons in an in vitro gastrointestinal model. Environ Sci Technol. 2001;45:1127–32. https://doi.org/10.1021/es1025849.

    Article  CAS  Google Scholar 

  19. Chen P, Stärkel P, Turner JR, Ho SB, Schnabl B. Dysbiosis-induced intestinal inflammation activates tumor necrosis factor receptor i and mediates alcoholic liver disease in mice. Hepatology. 2015;61:883–94.

    Article  CAS  PubMed  Google Scholar 

  20. Kiesslich R, Duckworth CA, Moussata D, Gloeckner A, Lim LG, Goetz M, et al. Local barrier dysfunction identified by confocal laser endomicroscopy predicts relapse in inflammatory bowel disease. Gut. 2012;61:1146–53. https://doi.org/10.1136/gutjnl-2011-300695.

    Article  CAS  PubMed  Google Scholar 

  21. Jacob RH, Afify AS, Shanab SM, Shalaby EA. Chelated amino acids: Biomass sources, preparation, properties, and biological activities. Biomass Convers Biorefin. 2022;14:2907–21. https://doi.org/10.1007/s13399-022-02333-3.

    Article  CAS  Google Scholar 

  22. Park HY, Hertz-Picciotto I, Petrik J, Palkovicova L, Kocan A, Trnovec T. Prenatal PBC exposure and thymus size at birth in neonates in eastern slovakia. Environ Health Perspect. 2008;116:104–9. https://doi.org/10.1289/ehp.9769.

    Article  CAS  PubMed  Google Scholar 

  23. Ogbuewu IP, Mbajiorgu CA. Potentials of dietary zinc supplementation in improving growth performance, health status, and meat quality of broiler chickens. Biol Trace Elem Res. 2023;201:1418–31. https://doi.org/10.1007/s12011-022-03223-5.

    Article  CAS  PubMed  Google Scholar 

  24. Alian HA, Samy HM, Ibrahim MT, Yusuf MS, Mahmoud MMA. Nano zinc oxide improves performance, IGF-1 mrna expression, meat quality, and humeral immune response and alleviates oxidative stress and NF-κB immunohistochemistry of broiler chickens. Biol Trace Elem Res. 2022;201:4062–78. https://doi.org/10.1007/s12011-022-03494-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lin PH, Sermersheim M, Li HC, Lee PHU, Steinberg SM, Ma JJ. Zinc in wound healing modulation. Nutrients. 2018;10:16–35. https://doi.org/10.3390/nu10010016.

    Article  CAS  Google Scholar 

  26. Weng X, Monteiro APA, Guo J, Li C, Orellana RM, Marins TN, et al. Effects of heat stress and dietary zinc source on performance and mammary epithelial integrity of lactating dairy cows. J Dairy Sci. 2018;101:2617–30. https://doi.org/10.3168/jds.2017-13484.

    Article  CAS  PubMed  Google Scholar 

  27. Shah M, Zaneb H, Masood S, Khan RU, Mobashar M, Khan I, et al. Single or combined applications of zinc and multi-strain probiotic on intestinal histomorphology of broilers under cyclic heat stress. Probiotics Antimicro. 2020;12:473–80. https://doi.org/10.1007/s12602-019-09561-6.

    Article  CAS  Google Scholar 

  28. Abuajamieh M, Abdelqader A, Irshaid R, Hayajneh FMF, Al-Khaza’leh JM, Al-Fataftah AR. Effects of organic zinc on the performance and gut integrity of broilers under heat stress conditions. Arch Anim Breed. 2020;63:125–35. https://doi.org/10.5194/aab-63-125-2020.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Bortoluzzi C, Lumpkins B, Mathis GF, Franca M, King WD, Graugnard DE, et al. Zinc source modulates intestinal inflammation and intestinal integrity of broiler chickens challenged with coccidia and clostridium perfringens. Poult Sci. 2019;98:2211–9. https://doi.org/10.3382/ps/pey587.

    Article  CAS  PubMed  Google Scholar 

  30. Zhang BK, Guo YM. Supplemental zinc reduced intestinal permeability by enhancing occludin and zonula occludens protein-1 (ZO-1) expression in weaning piglets. Br J Nutr. 2009;102:687–93. https://doi.org/10.1017/S0007114509289033.

    Article  CAS  PubMed  Google Scholar 

  31. Yan YW, Fan J, Bai SL, Hou WJ, Li X, Tong H. Zinc prevents abdominal aortic aneurysm formation by induction of A20-mediated suppression of NF-κB pathway. PLoS ONE. 2016;11: e0148536. https://doi.org/10.1371/journal.pone.0148536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Huang YL, Lu L, Li SF, Luo XG, Liu B. Relative bioavailabilities of organic zinc sources with different chelation strengths for broilers fed a conventional corn-soybean meal diet. J Anim Sci. 2009;87:2038–46. https://doi.org/10.2527/jas.2008-1212.

    Article  CAS  PubMed  Google Scholar 

  33. Huang YL, Lu L, Xie JJ, Li SF, Li XL, Liu SB, et al. Relative bioavailabilities of organic zinc sources with different chelation strengths for broilers fed diets with low or high phytate content. Anim Feed Sci Technol. 2013;179:144–8. https://doi.org/10.1016/j.anifeedsci.2012.10.010.

    Article  CAS  Google Scholar 

  34. Yu Y, Lu L, Wang RL, Xi L, Luo XG, Liu B. Effects of zinc source and phytate on zinc absorption by in situ ligated intestinal loops of broilers. Poult Sci. 2010;89:2157–65. https://doi.org/10.3382/ps.2009-00486.

    Article  CAS  PubMed  Google Scholar 

  35. Hu Y, Huang YH, Wang CL, Zhang WY, Qu YC, Li D, et al. The organic zinc with moderate chelation strength enhances the expression of related transporters in the jejunum and ileum of broilers. Poult Sci. 2023;102:102477. https://doi.org/10.1016/j.psj.2023.102477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Santos RR, Awati A, Roubos-van den Hil PJ, Tersteeg-Zijderveld MH, Koolmees PA, Fink-Gremmels J. Quantitative histo-morphometric analysis of heat-stress-related damage in the small intestines of broiler chickens. Avian Pathol. 2015;44:19–22. https://doi.org/10.1080/03079457.2014.988122.

    Article  CAS  PubMed  Google Scholar 

  37. Liu XX, Li HR, Lu A, Zhong YG, Hou XL, Wang N, et al. Reduction of intestinal mucosal immune function in heat-stressed rats and bacterial translocation. Int J Hyperther. 2012;28:756–65. https://doi.org/10.3109/02656736.2012.729173.

    Article  CAS  Google Scholar 

  38. Hu YY, Yang K, Zhang WY, Xue MX, Li TT, Wang SC, et al. Dietary Zn deficiency inhibits cell proliferation via the GPR39-mediated suppression of the PI3K/AKT/mTOR signaling pathway in the jejunum of broilers. Animals. 2024;14:979–89. https://doi.org/10.3390/ani14060979.

    Article  PubMed  PubMed Central  Google Scholar 

  39. NRC. Nutrient requirements of poultry (9th ed.). Washington, DC: The national academies press; 1994.

    Google Scholar 

  40. Ministry of Agriculture and Rural Affairs of the People's Republic of China. NY/T 33–2004 Feeding standard of chicken. Beijing: Agricultural press of China; 2004.

  41. Aviagen. Broiler management guide. Aviagen group: Midlothian, UK; 2009. Arbor Acres broiler: nutrition specification. https://aviagen.com/ap/brands/arbor-acres.

  42. Huang YL, Lu L, Luo XG, Liu B. An optimal dietary zinc level of broiler chicks fed a corn-soybean meal diet. Poult Sci. 2007;86:2582–9. https://doi.org/10.3382/ps.2007-00088.

    Article  CAS  PubMed  Google Scholar 

  43. Liao XD, Li A, Lu L, Liu SB, Li SF, Zhang LY, et al. Optimal dietary zinc levels of broiler chicks fed a corn–soybean meal diet from 22 to 42 days of age. Anim Prod Sci. 2013;53:388–94. https://doi.org/10.1071/an12291.

    Article  CAS  Google Scholar 

  44. Holwerda RA, Albin RC, Madsen FC. Chelation effectiveness of zinc proteinates demonstrated. Feedstuffs. 1995;67:12–3.

    Google Scholar 

  45. Tabler TW, Greene ES, Orlowski SK, Hiltz JZ, Anthony NB, Dridi S. Intestinal barrier integrity in heat-stressed modern broilers and their ancestor wild jungle fowl. Front Vet Sci. 2020;7:249–60. https://doi.org/10.3389/fvets.2020.00249.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Devita L, Nurilmala M, Lioe HAU, Suhartono MT. Chemical and antioxidant characteristics of skin-derived collagen obtained by acid-enzymatic hydrolysis of bigeye tuna (Thunnus obesus). Mar Drugs. 2021;19:222–40. https://doi.org/10.3390/md19040222.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang SC, Wu BX, Zhu L, Zhang WY, Zhang LY, Wu W, et al. The chemical characteristics of different sodium iron ethylenediaminetetraacetate sources and their relative bioavailabilities for broilers fed with a conventional corn-soybean meal diet. J Anim Sci Biotechnol. 2024;15:16. https://doi.org/10.1186/s40104-023-00969-3.

  48. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2-CT method. Methods. 2001;25:402–8. https://doi.org/10.1006/meth.2001.1262.

    Article  CAS  PubMed  Google Scholar 

  49. Liao XD, Liu GQ, Sun GM, Sun XM, Liu T, Lu L, et al. Determination of optimal dietary selenium levels by full expression of selenoproteins in various tissues of broilers from 1 to 21 d of age. Anim Nutr. 2021;7:1133–44. https://doi.org/10.1016/j.aninu.2021.02.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lu Z, He XF, Ma BB, Zhang L, Li JL, Jiang Y, et al. The alleviative effects and related mechanisms of taurine supplementation on growth performance and carcass characteristics in broilers exposed to chronic heat stress. Poult Sci. 2019;98:878–86. https://doi.org/10.3382/ps/pey433.

    Article  CAS  PubMed  Google Scholar 

  51. Arcila JCP, Tinoco IDFF, Saraz JAO, Rocha KSO, Candido MGL. Zootechnical and physiological performance of broilers in the final stage of growth subjected to different levels of heat stress. Rev Fac Agron. 2018;71:8469–76. https://doi.org/10.15446/rfna.v71n2.71927.

    Article  Google Scholar 

  52. He SJ, Yin QR, Xiong YJ, Li J, Liu DY. Characterization of heat stress affecting the growth performance, blood biochemical profile, and redox status in male and female broilers at market age. Trop Anim Health Prod. 2020;52:3833–41. https://doi.org/10.1007/s11250-020-02422-3.

    Article  PubMed  Google Scholar 

  53. Saleh AA, Ragab MM, Ahmed EAM, Abudabos AM, Ebeid TA. Effect of dietary zinc-methionine supplementation on growth performance, nutrient utilization, antioxidative properties and immune response in broiler chickens under high ambient temperature. J Appl Anim Res. 2017;46:820–7. https://doi.org/10.1080/09712119.2017.1407768.

    Article  CAS  Google Scholar 

  54. Lai PW, Liang JB, Hsia LC, Loh TC, Ho YW. Effects of varying dietary zinc levels and environmental temperatures on the growth performance, feathering score and feather mineral concentrations of broiler chicks. Austral J Anim Sci. 2010;23:937–45. https://doi.org/10.5713/ajas.2010.90495.

    Article  CAS  Google Scholar 

  55. Kuter E, Cengiz Ö, Köksal BH, Sevim Ö, Tatlı O, Ahsan U, et al. Litter quality and incidence and severity of footpad dermatitis in heat stressed broiler chickens fed supplemental zinc. Livest Sci. 2023;267:105145. https://doi.org/10.1016/j.livsci.2022.105145.

    Article  Google Scholar 

  56. Bartlett JR, Smith MO. Effects of different levels of zinc on the performance and immunocompetence of broilers under heat stress. Poult Sci. 2003;82:1580–8. https://doi.org/10.1093/ps/82.10.1580.

    Article  CAS  PubMed  Google Scholar 

  57. Zakaria HA, Jalal M, Al-Titi HH, Souad A. Effect of sources and levels of dietary zinc on the performance, carcass traits and blood parameters of broilers. Braz J Poultry Sci. 2017;19:519–26. https://doi.org/10.1590/1806-9061-2016-0415.

    Article  Google Scholar 

  58. Suttle NF. The mineral nutrition of livestock. 4th ed. Oxfordshire, UK: CABI publishing; 2010.

    Book  Google Scholar 

  59. Alhenaky A, Abdelqader A, Abuajamieh M, Al-Fataftah AR. The effect of heat stress on intestinal integrity and salmonella invasion in broiler birds. J Therm Biol. 2017;70:9–14. https://doi.org/10.1016/j.jtherbio.2017.10.015.

    Article  CAS  PubMed  Google Scholar 

  60. Nanto-Hara F, Kikusato M, Ohwada S, Toyomizu M. Heat stress directly affects intestinal integrity in broiler chickens. J Poult Sci. 2020;57:284–90. https://doi.org/10.2141/jpsa.0190004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Sun JJ, Zhang C, Zhang BK. Research note: Effects of organic zinc on broiler intestinal permeability and integrity in Clostridium perfringens–challenged condition. Poult Sci. 2020;99:6653–6. https://doi.org/10.1016/j.psj.2020.09.032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yan Y, Kolachala V, Dalmasso G, Nguyen H, Laroui H, Sitaraman SV, et al. Temporal and spatial analysis of clinical and molecular parameters in dextran sodium sulfate induced colitis. PLoS ONE. 2009;4:e6073. https://doi.org/10.1371/journal.pone.0006073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Senas-Cuesta R, Stein A, Latorre JD, Maynard CJ, Hernandez-Velasco X, Petrone-Garcia V, et al. The effects of essential oil from lippia origanoides and herbal betaine on performance, intestinal integrity, bone mineralization and meat quality in broiler chickens subjected to cyclic heat stress. Front Physiol. 2023;14:1184636. https://doi.org/10.3389/fphys.2023.1184636.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Zhang L, Gui SQ, Xu YH, Zeng JL, Wang J, Chen QR, et al. Colon tissue-accumulating mesoporous carbon nanoparticles loaded with musca domestica cecropin for ulcerative colitis therapy. Theranostics. 2021;11:3417–38. https://doi.org/10.7150/thno.53105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chu YX, Wang WH, Dai Y, Teng GG, Wang SJ. Esophageal helicobacter pylori colonization aggravates esophageal injury caused by reflux. World J Gastroenterol. 2014;20:15715–26. https://doi.org/10.3748/wjg.v20.i42.15715.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Lambert GP, Gisolfi CV, Berg DJ, Moseley PL, Oberley LW, Kregel KC. Molecular biology of thermoregulation selected contribution: Hyperthermia-induced intestinal permeability and the role of oxidative and nitrosative stress. J Appl Physiol. 2002;92:1750–61. https://doi.org/10.1152/japplphysiol.00787.2001.

    Article  CAS  PubMed  Google Scholar 

  67. Lan RX, Li YX, Chang QQ, Zhao ZH. Dietary chitosan oligosaccharides alleviate heat stress-induced intestinal oxidative stress and inflammatory response in yellow-feather broilers. Poult Sci. 2020;99:6745–52. https://doi.org/10.1016/j.psj.2020.09.050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Pearce SC, Fernandez MVS, Torrison J, Wilson ME, Baumgard LH, Gable NK. Dietary organic zinc attenuates heat stress–induced changes in pig intestinal integrity and metabolism. J Anim Sci. 2015;93:4702–13. https://doi.org/10.2527/jas2015-9018.

    Article  CAS  PubMed  Google Scholar 

  69. Burin Junior AM. Intestinal integrity, oxidative stress, and immune competence of broilers exposed to heat stress and supplemented with Zn amino acid complex. Doctoral Thesis. Escola Superior de Agricultura Luiz de Queiroz. Piracicaba, Brazil: University of São Paulo; 2019. p. 33–53.

  70. Chang YQ, Tang HY, Zhang ZY, Yang T, Wu B, Zhao H, et al. Zinc methionine improves the growth performance of meat ducks by enhancing the antioxidant capacity and intestinal barrier function. Front Vet Sci. 2022;9:774160. https://doi.org/10.3389/fvets.2022.774160.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Shah M, Zaneb H, Masood S, Khan I, Sikandar A, Ashraf S, et al. Effect of zinc and probiotics supplementation on performance and immune organs morphology in heat stressed broilers. S Afr J Anim Sci. 2019;48:1017–25. https://doi.org/10.4314/sajas.v48i6.3.

    Article  CAS  Google Scholar 

  72. Sanz Fernandez MV, Pearce SC, Gabler NK, Patience JF, Wilson ME, Socha MT, et al. Effects of supplemental zinc amino acid complex on gut integrity in heat-stressed growing pigs. Animal. 2014;8:43–50. https://doi.org/10.1017/S1751731113001961.

    Article  CAS  PubMed  Google Scholar 

  73. Xu ZAO, Li DQ, Qu W, Yin YX, Qiao SP, Zhu YN, et al. Card9 protects sepsis by regulating Ripk2-mediated activation of NLRP3 inflammasome in macrophages. Cell Death Dis. 2022;13:502–13. https://doi.org/10.1038/s41419-022-04938-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ruff J, Barros TL, Tellez G Jr, Blankenship J, Lester H, Graham BD, et al. Research note: Evaluation of a heat stress model to induce gastrointestinal leakage in broiler chickens. Poult Sci. 2020;99:1687–92. https://doi.org/10.1016/j.psj.2019.10.075.

    Article  PubMed  Google Scholar 

  75. Siddiqui SH, Kang D, Park J, Khan MS, Shim K. Chronic heat stress regulates the relation between heat shock protein and immunity in broiler small intestine. Sci Rep. 2020;10:18872. https://doi.org/10.1038/s41598-020-75885-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Xia B, Wu WD, Fang W, Wen XB, Xie JJ, Zhang HF. Heat stress-induced mucosal barrier dysfunction is potentially associated with gut microbiota dysbiosis in pigs. Anim Nutr. 2022;8:289–99. https://doi.org/10.1016/j.aninu.2021.05.012.

    Article  CAS  PubMed  Google Scholar 

  77. Xie YQ, Wen M, Zhao H, Liu GM, Chen XL, Tian G, et al. Effect of zinc supplementation on growth performance, intestinal development, and intestinal barrier function in pekin ducks with lipopolysaccharide challenge. Poult Sci. 2021;100:101462. https://doi.org/10.1016/j.psj.2021.101462.

  78. Al-Sadi R, Ye D, Said HM, Ma TY. IL-1 beta-induced increase in intestinal epithelial tight junction permeability is mediated by MEKK-1 activation of canonical NF-κB pathway. Am J Pathol. 2010;177:2310–22. https://doi.org/10.2353/ajpath.2010.100371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhu KZ, Liang W, Ma ZJ, Xu DC, Cao SY, Lu XJ, et al. Necroptosis promotes cell-autonomous activation of proinflammatory cytokine gene expression. Cell Death Dis. 2018;9:500–15. https://doi.org/10.1038/s41419-018-0524-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Ma JJ, Shi M, Zhang XC, Liu XN, Chen J, Zhang RD, et al. GLP-1R agonists ameliorate peripheral nerve dysfunction and inflammation via p38 MAPK/NF-κB signaling pathways in streptozotocin-induced diabetic rats. Int J Mol Med. 2018;41:2977–85. https://doi.org/10.3892/ijmm.2018.3509.

    Article  CAS  PubMed  Google Scholar 

  81. Cheng WT, Wang N. Correlation between MMP-2 and NF-κB expression of intracranial aneurysm. Asian Pac J Trop Med. 2013;6:570–3. https://doi.org/10.1016/s1995-7645(13)60098-x.

    Article  CAS  PubMed  Google Scholar 

  82. Bond M, Fabunmi RP, Baker AH, Newby AC. Synergistic upregulation of metalloproteinase-9 by growth factors and inflammatory cytokines: An absolute requirement for transcription factor NF-κB. FEBS Lett. 1998;435:29–34. https://doi.org/10.1016/s0014-5793(98)01034-5.

    Article  CAS  PubMed  Google Scholar 

  83. Takahashi M, Terada Y, Nakai I, Nakanishi H, Yoshimura E, Mori S, et al. Role of nicotianamine in the intracellular delivery of metals and plant reproductive development. Plant Cell. 2003;15:1263–80. https://doi.org/10.1105/tpc.010256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Catrysse L, Vereecke L, Beyaert R, Loo Gv. A20 in inflammation and autoimmunity. Trends Immunol. 2014;35:22–31. https://doi.org/10.1016/j.it.2013.10.005.

    Article  CAS  PubMed  Google Scholar 

  85. Li CW, Guo SS, Gao J, Guo YM, Du EC, Lv ZP, et al. Maternal high-zinc diet attenuates intestinal inflammation by reducing DNA methylation and elevating H3K9 acetylation in the A20 promoter of offspring chicks. J Nutr Biochem. 2015;26:173–83. https://doi.org/10.1016/j.jnutbio.2014.10.005.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Key International Cooperation Program of the National Natural Science Foundation of China (32120103011), the Jiangsu Shuang Chuang Tuan Dui program (JSSCTD202147), the Jiangsu Shuang Chuang Ren Cai program (JSSCRC2021541) and the Initiation Funds of Yangzhou University for Distinguished Scientists.

Author information

Authors and Affiliations

Authors

Contributions

We thank all authors for their contributions to the present study. Both XGL and YH were responsible for all issues related to this paper. YYH was responsible for animal experiment, sample collections and analyses, and statistical analyses of all data, as well as the manuscript writing. WYZ, KY, TTL, SCW, XYC, XDL, LYZ and SFL were involved in sample collections and analyses. XL, HCL, JO and MTS were involved in the experimental design, data interpretations and manuscript corrections. All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Yun Hu or Xugang Luo.

Ethics declarations

Ethics approval and consent to participate

The experiment was approved by the Animal Ethics Committee of Yangzhou University (approval number: SYXK (Su) 2021–0027).

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest.

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

Hu, Y., Zhang, W., Yang, K. et al. Dietary Zn proteinate with moderate chelation strength alleviates heat stress-induced intestinal barrier function damage by promoting expression of tight junction proteins via the A20/NF-κB p65/MMP-2 pathway in the jejunum of broilers. J Animal Sci Biotechnol 15, 115 (2024). https://doi.org/10.1186/s40104-024-01075-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-024-01075-8

Keywords