Next Article in Journal
Protection of Chickens against H9N2 Avian Influenza Isolates with a Live Vector Vaccine Expressing Influenza Hemagglutinin Gene Derived from Y280 Avian Influenza Virus
Next Article in Special Issue
Dietary Peppermint Extract Inhibits Chronic Heat Stress-Induced Activation of Innate Immunity and Inflammatory Response in the Spleen of Broiler Chickens
Previous Article in Journal
Fetal Programming Influence on Microbiome Diversity and Ruminal and Cecal Epithelium in Beef Cattle
Previous Article in Special Issue
Impact of Phytase Supplementation on Meat Quality of Heat-Stressed Broilers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Dietary Hydroxytyrosol Supplementation on Growth Performance, Gut Morphometry, and Oxidative and Inflammatory Status in LPS-Challenged Broilers

1
Department of Animal Science, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
2
Nova Mentis Limited, Nova UCD, Belfield Innovation Park, University College Dublin, Belfield, D04 F438 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Deceased author.
Animals 2024, 14(6), 871; https://doi.org/10.3390/ani14060871
Submission received: 29 January 2024 / Revised: 5 March 2024 / Accepted: 11 March 2024 / Published: 12 March 2024
(This article belongs to the Special Issue Second Edition of Stress Management in Poultry)

Abstract

:

Simple Summary

Broiler chickens are frequently exposed to a variety of stress factors in the poultry industry. To simulate these factors, lipopolysaccharide (LPS) from E. coli is commonly used as a research model to stimulate inflammatory and oxidative stresses in broilers. LPS can reduce growth performance parameters by negatively affecting broilers’ immune response to inflammation. Feed additives that could mitigate those detrimental effects are often demanded. The present study evaluated the impact of hydroxytyrosol supplementation on the growth performance, gut morphometry, and anti-inflammatory and antioxidant parameters of broilers challenged with LPS. Hydroxytyrosol improved the growth performance, gut morphometry, and oxidative status of broilers challenged with LPS. Therefore, hydroxytyrosol supplementation is a powerful nutritional intervention for mitigating the detrimental effects of LPS.

Abstract

This study assessed the effects of hydroxytyrosol (HT) on 8- to 20-day-old broilers challenged with lipopolysaccharide (LPS); 180 Cobb500™ male chicks were randomly assigned to 3 treatment groups, each comprising 10 replicates with 6 birds per replicate. Treatments included a control diet (CON), CON with LPS administration, and CON + LPS supplemented with 10 mg of HT/kg of feed. LPS was administered intraperitoneally on days 14, 16, 18, and 20. Body weight (BW), body weight gain (BWG), and the feed conversion ratio (FCR) were measured. On day 20, ten birds per treatment were slaughtered for analysis. Bursa, spleen, and liver were collected, and their respective relative weight was determined. The jejunum was destined for morphological analyses of villus height (VH), crypt depth (CD), and their ratio (VH:CD), and for mRNA expression of nuclear factor kappa B (NF-κB), catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), and interleukins 10 (IL-10), 1 beta (IL-1β), and 8 (IL-8). HT improved BW, BWG, and FCR, and reduced crypt depth (CD) while increasing the VH:CD ratio in the jejunum. Moreover, HT downregulated mRNA expression of CAT, GPx, IL-10, and IL-1β. In conclusion, HT enhances broiler growth performance, mitigates jejunal mucosa damage from LPS, and modulates antioxidant and immune responses.

1. Introduction

In the poultry industry, broilers are frequently exposed to a variety of stress situations such as microbially contaminated feed, pathogen infection, and immunological and oxidative stresses contributing to intestinal mucosa injury and reduction of growth performance [1,2,3]. Lipopolysaccharide (LPS), an integral component of the cell wall found in Gram-negative bacteria, serves as a standard inducer of inflammatory and oxidative stresses in broilers [4,5]. LPS-challenged broilers present an acute inflammatory response and oxidative damage, such as a reduction in the expression and activity of antioxidant enzymes, elevated production of pro-inflammatory cytokines, increased weight of lymphoid organs, and impaired digestion and absorption of nutrients due to morphological damage in the intestine [1,3,6,7].
Plant polyphenols such as curcumin, resveratrol, and hydroxytyrosol (HT) have potential health effects. They are considered potent immunomodulators with many physiological effects due to their antioxidant and anti-inflammatory activities, including for poultry health [8,9,10,11]. HT is found in olives and has been shown to confer many health benefits [8]. It is one of the most powerful naturally known antioxidants and its antioxidant capacity can be attributed to its ortho-dihydroxy configuration within its aromatic ring. According to Bertelli et al. [12], HT’s antioxidant capacity surpasses green tea by tenfold and coenzyme Q10 by twice the amount. Moreover, HT is acknowledged for its role as a scavenger of reactive oxygen species (ROS). HT and its metabolites can reduce the intracellular and extracellular accumulation of ROS and protect vascular endothelial cells from hydrogen peroxide [13,14,15]. Other beneficial effects of HT include the promotion of immunity, anti-inflammatory effects, and the promotion of gut health [7,12,16,17], but previous studies with HT have not shown an increase in broiler growth performance [18,19,20].
No studies have been carried out on chickens exposed to LPS and HT. Consequently, there is a lack of knowledge and understanding of how HT influences the immune response, intestinal health, and growth performance of LPS-challenged broiler chickens. We hypothesized that HT has an anti-inflammatory effect, and its dietary supplementation could mitigate the detrimental effects of LPS challenge on broilers’ growth performance. Therefore, this research aimed to evaluate the effects of HT on immune and antioxidant responses, as well as intestinal mucosa regulation through measurement of growth performance, morphological changes in the jejunum, and gene expression of antioxidant enzymes and inflammatory parameters.

2. Materials and Methods

2.1. Animals, Experimental Design, and Diets

The study was conducted at the Unit of Study and Research in Poultry Production and Nutrition of the Department of Animal Science, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil.
Cobb500™ male broiler chicks were purchased from a commercial hatchery (Rivelli Alimentos SA, Matheus Leme, MG, Brazil). At the hatchery, all chicks were vaccinated against Marek and Newcastle diseases and infectious bronchitis. Before reaching 8 d, broiler chicks were reared in accordance with the guidelines outlined by Cobb500®. During this period, they had unrestricted access to water and a diet formulated with corn and soybean meal under the guidelines established by Rostagno et al. [21].
Following, 180 Cobb500™ male chicks (230.9 g ± 3.44 g) were accommodated in metallic cages (600 cm2/bird) and assigned randomly to 3 treatment groups with 10 replicates of 6 birds each, from 8 to 20 days old. Every cage was equipped with a nipple drinker and a trough feeder, ensuring unrestricted access to water and feed throughout the experimental period. The temperature was maintained at 28 °C at the beginning of the experiment, and it was gradually reduced to 22 °C by 20 days of age. Birds were daily exposed to 18 h of continuous light during the experimental period.
The treatments consisted of a control diet (CON), a CON with LPS administration (CON + LPS), and a CON supplemented with 10 mg of hydroxytyrosol/kg of feed with LPS administration (HT + LPS). The experimental diets were formulated based on corn and soybean meal to fulfill the nutritional requirements of the birds according to the guidelines provided by Rostagno et al. [21], without the addition of antibiotics or anticoccidial (Table 1). 1-HT® hydroxytyrosol (>98% purity) used in this study was biologically manufactured and provided by Nova Mentis Ltd., Dublin, Ireland.
Escherichia coli LPS (serotype O55:B5, Sigma Chemical Co., St. Louis, MO, USA) was reconstituted in saline solution at a concentration of 1.0 mg/mL and administered to the birds intraperitoneally at a dose of 1 mL/kg of body weight on days 14, 16, 18, and 20. Birds from the control group received the same amount of saline solution (0.9% sodium chloride) via intraperitoneal to guarantee similar handling and stress conditions induced in the ones that received LPS administration.

2.2. Growth Performance

At the start and end of the experimental period, which occurred at 8 and 20 days, respectively, the birds and their feed were weighed. Data were collected to measure body weight (BW, kg/bird), BW gain (BWG, kg/bird), feed intake (FI, kg/bird), and feed conversion ratio (FCR, kg/kg). In cases of mortality, the remaining feed was weighed to adjust the FI measurement.

2.3. Samples Collection

At 20 days old, one bird with BW closest to the average weight of the experimental unit was chosen for sample collection (totaling 10 birds per treatment). Four hours after the first LPS application, the bird was slaughtered by cervical dislocation for sample collection. Jejunum was identified using Meckel’s diverticulum as a reference, and aseptically removed and separated into two different samples to evaluate intestinal morphometry and determine mRNA content. Furthermore, the liver, spleen, and bursa were removed to determine their relative weights.

2.3.1. Relative Organ Weights

Before slaughtering, the birds selected were weighed. After slaughtering and evisceration, the liver, spleen, and bursa were removed and weighed. Relative organ weight (ROW) was calculated in relation to the birds’ live weight.

2.3.2. Intestinal Morphometry

Samples of the jejunum (≈3 cm length) were obtained and rinsed using a sterile saline solution (0.9% sodium chloride) to clear any luminal contents. The segments were longitudinally incised and affixed to the cardboard using staples, where the serosa remained in contact with the cardboard to not distend or harm its structures [22]. Samples were labeled and preserved in sterile plastic containers filled with a 10% buffered formalin solution. Afterward, the samples underwent dehydration by immersing them in a series of increasing alcohol concentrations, followed by clarification in xylene, and finally embedding in liquid paraffin at 60 °C. The segments were submitted to microtomy to produce semi-serial cross-sections with a thickness of 5 µm. These sections were then stained with hematoxylin and eosin, following Luna’s method [23]. Five 5 µm thick cross-sections were placed on every microscope slide. The slides were observed and photographed using an optical microscope (EVOS® XL Core) with 10× magnification. Subsequently, measurements of villus heights (VH), crypt depths (CD), and the villus height to crypt depth ratio (VH:CD) were performed using ImageJ 1.50i Software, java1.6.0_20 (National Institutes of Health, Bethesda, MD, USA). A total of 20 villi and their crypt were measured for each experimental unit (200 villi and crypt per treatment).

2.3.3. mRNA Extraction and Gene Expression

A second sterile portion of jejunum was obtained, individually stored in cryogenic tubes, and preserved in liquid nitrogen until further analysis. Total RNA extraction was conducted by employing Trizol® (Invitrogen, Carlsbad, CA, USA) following the manufacturer’s instructions. RNase-free silica membrane columns (PureLink™ RNA Mini Kit—Invitrogen TM) were used to wash and isolate the RNA. The resulting precipitate was rehydrated using 30 µL of UltraPure® DNase/RNase-Free water. The RNA concentration was estimated using a NanoDrop™ Lite spectrophotometer (Thermo Fisher Scientific, Beverly, MA, USA) with purity verified by observing A260/A280 ratios within the range of 1.8 to 2.0. RNA integrity was assessed on a 1% agarose gel. Subsequently, the samples underwent DNase treatment and were reverse transcribed into cDNA using the High-Capacity cDNA Reverse Transcription Kits (Applied Biosystems, Thermo Fisher Scientific, Beverly, MA, USA) according to the manufacturer’s instructions.
The target genes evaluated in jejunum were nuclear factor kappa B (NF-κB), catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD1), and interleukins 1β (IL-1β), 10 (IL-10), and 8 (IL-8). β-actin (β-ACT) was chosen as the endogenous gene for data normalization because of its high expression level and stability (Table 2).
The real-time quantitative PCR (RT-qPCR) analyses were conducted in duplicate on a QuantStudio 3 Thermocycler (Applied Biosystems™, Foster City, CA, USA), utilizing the Relative Quantification approach. Detection was achieved with the SYBR® Green system (Applied Biosystems, Foster City, CA, USA), and the GoTaq® qPCR Master Mix kit (Promega Corporation, Madison, WI, USA) was used. The PCR followed a specific cycling protocol: an initial denaturation at 95 °C for 2 min, followed by 40 amplification cycles of denaturation at 95 °C for 15 s each, and an annealing and extension phase at 60 °C for 1 min. After amplification, the threshold cycle (Ct) values were normalized employing the ΔCt method, comparing against Ct values derived from the endogenous control gene β-ACT. The quantification of relative gene expression levels was performed employing the 2−ΔCt method as elucidated by Livak and Schmittgen [24].

2.4. Statistical Analysis

Analysis of the data was conducted using one-way ANOVA executed through the ExpDes.pt package in R statistical software (version 4.0.4), with the Tukey test applied for mean comparison. A significance level was set at α = 0.05, and each replicate was regarded as an independent experimental unit.

3. Results

3.1. Growth Performance

The birds that received the LPS challenge without HT supplementation (CON + LPS) showed worse BWG, BW, and FCR, compared to the CON and HT + LPS groups (p < 0.001) (Table 3). There were no differences between the treatments on FI (p = 0.09).

3.2. Relative Organ Weight

Birds challenged with LPS, with or without HT supplementation, had an increase in liver ROW (p = 0.001) and in spleen ROW (p < 0.0001) compared to CON (Table 4). There were no differences between the treatments for Bursa ROW (p = 0.997).

3.3. Intestinal Morphometry

Birds from CON + LPS showed an increase in CD (p < 0.001) and decreased the VH:CD ratio of jejunum (p = 0.004) when compared to birds in the CON and HT + LPS treatment groups (Table 5). However, there was no difference in VH among the treatment groups (p = 0.324). To visualize the effects, images can be found in Figure S1.

3.4. mRNA Expression

Birds from the HT + LPS treatment group showed lower gene expression of CAT (p = 0.03) and GPx (p = 0.0004) when compared to CON (Figure 1A,B). Birds challenged with LPS but without dietary HT showed higher gene expression for IL-10 (p < 0.0001) (Figure 1C). Furthermore, birds treated with HT + LPS had lower gene expression of IL-1β compared to CON (p = 0.022) (Figure 1D). No significant differences were observed in the gene expression levels of IL-8 (p = 0.213), NF-κB (p = 0.633), and SOD1 (p = 0.315) among the treatment groups (Figure 1E–G).

4. Discussion

Gram-negative bacteria have LPS, an endotoxin, as a main component of their cell wall. The release of LPS during bacterial death or rapid growth is associated with the release of pro-inflammatory cytokines and can be harmful to broiler development [25,26]. Previous studies showed that challenges with LPS affected feed intake and weight gain of broiler chickens [3,5,27]. Similarly, in the current study, LPS administration caused a reduction in BWG and BW and increased FCR. However, in the current study, when birds were fed dietary HT, those parameters were not negatively affected when compared to birds in the control treatment. In previous research, supplementation of HT had no influence on broiler growth performance when they were reared under normal conditions [18,19,20]. These previous findings together with the findings of the current study suggest that HT may modulate the inflammatory response, caused by LPS administration, and thus, decrease the diversion of nutrients required to support the immune system due to its anti-inflammatory activity [7], instead of directly promoting broiler growth performance.
The liver, spleen, and bursa of Fabricius play crucial roles in general metabolism and serve as immune organs in broilers. Also, they are LPS target organs, being relevant during the acute-phase immune response [6,28]. The ROW of these organs is a useful parameter to evaluate the immune function of poultry [3]. Several studies have shown that LPS administration caused an increase in the ROW of the liver and spleen, which may be explained by the increase in the production of pro-inflammatory cytokines and recruitment of inflammatory cells to these organs, resulting in its mass growth [6,29,30,31]. We observed that birds that received LPS administration had an increase in ROW of the liver and spleen, regardless of the use or non-use of HT in diets, when compared to birds from the control group. This suggests that dietary HT was not capable of reducing liver and spleen ROW.
The barrier formed by the intestinal mucosa is crucial for maintaining gut balance and acts as the initial defense line against pathogens, facilitated by the digestion and absorption of nutrients [32]. The small intestine is the primary site for nutrient absorption, but it is also a targeted organ for LPS [33,34]. Intestinal mucosa dysfunction is characterized by histological changes such as villus necrosis, reduced VH, and increased CD in the mucosa [35]. An increase in CD and a decrease in VH can impair nutrient absorption, increase the secretion of electrolytes and water in the gastrointestinal tract, and can consequently worsen broiler growth performance [36]. A deeper crypt may imply faster tissue turnover to facilitate the renewal of villi, indicating that the host’s intestinal response mechanism is attempting to compensate for regular sloughing or atrophy of the villus caused by inflammation from pathogens and their toxins [17,37]. Therefore, a high VH:CD ratio is considered a reliable marker of mucosal turnover and is linked to an enhanced capacity for digestion and nutrient absorption [38,39]. The results showed negative effects of LPS administration on intestinal morphometry, evidenced by increasing CD and decreasing VH:CD, consistent with previous findings [2,3,40]. However, dietary HT could attenuate intestinal morphometric damage by decreasing CD and increasing the VH:CD ratio, which consequently could be observed in increased broiler growth performance. These findings could be explained by HT’s role through a direct biological activity in the gastrointestinal tract before being absorbed and also acting on intestinal flora regulation, where HT improves microbial community disturbance and protects intestinal wall integrity [7,41].
LPS initiates the inflammatory process via a Toll-like receptor (TLR4), which is expressed on the cell membrane of leukocytes. After LPS activates TLR4, NF-κB accesses the nucleus and modulates the release of pro-inflammatory cytokines [42,43,44,45]. The interleukin IL-1β plays a pivotal role in regulating the innate immune response in birds [1]. In our study, the LPS challenge did not alter the gene expression of NF-κB and IL-8, while it was observed that the expression of IL-1β was lower in the LPS + HT treatment, compared to CON, but not different from CON + LPS. Therefore, the effects of HT as an anti-inflammatory agent could not be determined in the present study. Moreover, the expression of IL-10 decreased in the LPS + HT treatment when compared to the CON + LPS. IL-10 is known to be a potent anti-inflammatory cytokine that regulates the host immune response to minimize unintended host cell damage during inflammation [46]. The reduction in both interleukins was also observed by Kaiser et al. [47] for vitamin E and Zhang et al. [48] for α-tocopherol succinate supplementations. These results could be attributed to a mechanism of homeostasis in which the appropriate balance of the cytokine network is maintained uniformly for both pro- and anti-inflammatory cytokines.
The administration of LPS not only induces inflammation but also leads to oxidative stress, which is related to inflammatory responses by the release of pro-inflammatory cytokines [26,30,49]. The antioxidant system is composed mainly of SOD, CAT, and GPx, which act as a scavenger of free radicals to decompose hydrogen peroxide (H2O2) [2,32]. SOD acts on superoxide anion, which is dismutated to H2O2, and subsequently is catalyzed to H2O by CAT, GPx, or peroxiredoxins [50]. Interestingly, we found that dietary HT reduced the expression of GPx and CAT in the jejunum. It suggests that HT may act directly on H2O2, consequently reducing the need for GPx and CAT to catalyze it into H2O. Similar to our finding, Zrelli et al. [13] reported that HT had a protective effect on vascular endothelial cells against the cytotoxic effects of H2O2. The authors led an investigation to understand the mechanisms involved in this protection and discovered that HT acted through PI3K/Akt and ERK1/2 pathways. These enzymes often play a role in stimulating cell multiplication and enhancing cell survival during oxidative stress [13].
Since the findings showed that HT supplementation decreased antioxidant enzyme expressions, we hypothesized that HT could be acting through another pathway that was not investigated in the present study. The literature shows that HT can modulate the antioxidant response through the nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 is one of the major regulators of the antioxidant response and prevents oxidative damage [51]. It can regulate the expression of a diversity of genes related to antioxidant defense and detoxification. Under normal circumstances, Nrf2 is bound to a cytoplasmatic protein called KEAP1, which targets Nrf2 for ubiquitination and subsequent degradation, maintaining low levels of Nrf2 in the cell. On the other hand, under stress conditions, like the LPS challenge in the present study, KEAP1 undergoes a conformational change and thus, does not target Nrf2 for degradation [13,52,53]. Nrf2 activation occurs after its translocation into the nucleus, where it binds to the antioxidant response element (ARE) to promote heme oxygenase 1 (HO-1) and other antioxidant gene expression [13,51,52]. HT has been correlated with the ARE pathway, in which it promotes the activation of various genes encoding ARE, including DNA-repair proteins or phase II detoxifying enzymes [14]. In addition to the activation of PI3K/Akt and ERK1/2 pathways, HT induces the expression of HO-1, a downstream phase II detoxifying enzyme, after Nrf2 activation [13]. HO-1 reduces intracellular ROS, therefore, contributing to the cytoprotective effects of HT against oxidative stress [13]. According to Satta et al. [51], the Nrf2/HO-1 signaling pathway has a pivotal role in decreasing oxidative stress levels in the organism.
In the present study, HT acted by reducing the oxidative stress in the broiler’s jejunum. It could be related mainly to HT’s direct role against the cytotoxic effects of H2O2 as previously discussed. Moreover, it might have induced an increase in HO-1 gene expression, which acts to reduce intracellular ROS; however, it was not investigated.
Briefly, HT has an important role in the antioxidant response, in which the proper mechanisms need to be further investigated to understand HT’s antioxidant capacity either directly or indirectly.

5. Conclusions

The supplementation of 10 mg of HT/kg of feed attenuates the reduced growth performance and the jejunal mucosa damage caused by LPS challenge by increasing villus height to crypt depth ratio and improving the oxidative response of broiler chickens. More studies should be carried out to better elucidate if HT has anti-inflammatory effects on broilers and to understand the proper mechanisms by which HT can enhance the oxidative responses of broiler chickens.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ani14060871/s1, Figure S1: Images of jejunum to illustrate the effects of hydroxytyrosol supplementation on the morphology, collected from broiler chickens at 20 days old, challenged or not by LPS.

Author Contributions

Conceptualization, K.M.M.D., L.F.T.A., H.S.R., K.E.O., R.D., M.W., J.B. and E.A.A.; methodology, K.M.M.D., C.H.O., A.A.C., L.F.T.A. and H.S.R.; software, K.M.M.D. and C.H.O.; validation, K.M.M.D. and C.H.O.; formal analysis, K.M.M.D., C.H.O. and K.M.G.; investigation, K.M.M.D., C.H.O. and K.M.G.; resources, L.F.T.A. and E.A.A.; data curation, K.M.M.D. and C.H.O.; writing—original draft preparation, K.M.M.D.; writing—review and editing, C.H.O., A.A.C., H.S.R., K.E.O., R.D., M.W. and J.B.; visualization, K.M.M.D., L.F.T.A., A.A.C. and H.S.R.; supervision, L.F.T.A., A.A.C. and H.S.R.; project administration, L.F.T.A. and H.S.R.; funding acquisition, L.F.T.A. and H.S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The Ethics Committee in the Use of Farming Animals at the Federal University of Viçosa approved all procedures utilized in this research prior to implementation under protocol number 047/2022, approval date: 22 June 2022, following the guidelines of the National Council for Experimentation Animal Control.

Informed Consent Statement

The content of Table 1 was created by the authors based on the information available in the reference by Rostagno et al. [21]. Regarding the broiler chicks mentioned in the manuscript, they were purchased directly from a commercial hatchery (Rivelli Alimentos SA, Matheus Leme, MG, Brazil) and did not have individual owners. Therefore, there was no need to obtain written informed consent from owners or authorized agents.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

We dedicate this study to the memory of our beloved Luiz Albino, an extraordinary mentor, collaborator, and friend whose presence illuminated not only our professional but also our personal lives. His legacy will endure through the many minds he touched. We thank Nova Mentis Limited for providing their biotechnologically high-purity hydroxytyrosol and for supporting this work. This work was also supported by CNPQ—Conselho Nacional de Desenvolvimento Científico e Tecnológico and CAPES—Coordenação de Aperfeiçoamento de Pessoal de Nível Superior.

Conflicts of Interest

Kevin O. Connor and Enrico Altieri are founders of Nova Mentis, Ltd. Reeta Davis, Meg Walsh, and James Britton are employees of Nova Mentis, Ltd. Nova Mentis does not have any patents related to the use of hydroxytyrosol in animals. Nova Mentis produced hydroxytyrosol for this study, co-designed the animal feed experiments, and analyzed the data generated in the study. The authors declare no conflict of interest.

References

  1. Chen, J.; Wang, H.; Wu, Z.; Gu, H.; Li, C.; Wang, S.; Liu, G. Effects of 5-aminolevulinic acid on the inflammatory responses and antioxidative capacity in broiler chickens challenged with lipopolysaccharide. Animal 2022, 16, 100575. [Google Scholar] [CrossRef]
  2. Kong, L.; Wang, Z.; Xiao, C.; Zhu, Q.; Song, Z. Glycerol monolaurate attenuated immunological stress and intestinal mucosal injury by regulating the gut microbiota and activating AMPK/Nrf2 signaling pathway in lipopolysaccharide-challenged broilers. Anim. Nutr. 2022, 10, 347–359. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, Y.; Ye, J.; Zhang, S.; Chen, Z.; Fan, Q.; Jiang, S. Dietary supplementation with anthocyanin attenuates lipopolysaccharide-induced intestinal damage through antioxidant effects in yellow-feathered broiler chicks. Poult. Sci. 2022, 102, 102325. [Google Scholar] [CrossRef] [PubMed]
  4. Yang, L.; Liu, G.; Liang, X.; Wang, M.; Zhu, X.; Luo, Y.; Shang, Y.; Yang, J.-Q.; Zhou, P.; Gu, X.-L. Effects of berberine on the growth performance, antioxidative capacity and immune response to lipopolysaccharide challenge in broilers. Anim. Sci. J. 2019, 90, 1229–1238. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, J.; Han, H.; Zhang, L.; Wang, T. Dietary bisdemethoxycurcumin supplementation attenuates lipopolysaccharide-induced damages on intestinal redox potential and redox status of broilers. Poult. Sci. 2021, 100, 101061. [Google Scholar] [CrossRef] [PubMed]
  6. Bi, S.; Shao, J.; Qu, Y.; Hu, W.; Ma, Y.; Cao, L. Hepatic transcriptomics and metabolomics indicated pathways associated with immune stress of broilers induced by lipopolysaccharide. Poult. Sci. 2022, 101, 102199. [Google Scholar] [CrossRef] [PubMed]
  7. Shan, C.; Miao, F. Immunomodulatory and antioxidant effects of hydroxytyrosol in cyclophosphamide-induced immunosuppressed broilers. Poult. Sci. 2022, 101, 101516. [Google Scholar] [CrossRef] [PubMed]
  8. Britton, J.; Davis, R.; O’Connor, K.E. Chemical, physical and biotechnological approaches to the production of the potent antioxidant hydroxytyrosol. Appl. Microbiol. Biotechnol. 2019, 103, 5957–5974. [Google Scholar] [CrossRef] [PubMed]
  9. Zhang, J.; Sun, X.; Chai, X.; Jiao, Y.; Sun, J.; Wang, S.; Wang, S.; Yu, H.; Feng, X. Curcumin mitigates oxidative damage in broiler liver and ileum caused by Aflatoxin B1-contaminated feed through Nrf2 signaling pathway. Animals 2024, 14, 409. [Google Scholar] [CrossRef]
  10. Yang, H.; Wang, Y.; Liu, M.; Liu, X.; Jiao, Y.; Jin, S.; Shan, A.; Feng, X. Effects of dietary resveratrol supplementation on growth performance and anti-inflammatory ability in ducks (Anas platyrhynchos) through the Nrf2/HO-1 and TLR4/NF-κB signaling pathways. Animals 2021, 11, 3588. [Google Scholar] [CrossRef]
  11. Yang, H.; Wang, Y.; Jin, S.; Pang, Q.; Shan, A.; Feng, X. Dietary resveratrol alleviated lipopolysaccharide-inducted ileitis through Nrf2 and NK-κB signalling pathways in ducks (Anas platyrhynchos). J. Anim. Physiol. Anim. Nutr. 2022, 106, 1306–1320. [Google Scholar] [CrossRef]
  12. Bertelli, M.; Kiani, A.K.; Paolacci, S.; Manara, E.; Kurti, D.; Dhuli, K.; Bushati, V.; Miertus, J.; Pangallo, D.; Baglivo, M.; et al. Hydroxytyrosol: A natural compound with promising pharmacological activities. J. Biotechnol. 2020, 309, 29–33. [Google Scholar] [CrossRef] [PubMed]
  13. Zrelli, H.; Matsuoka, M.; Kitazaki, S.; Araki, M.; Kusunoki, M.; Zarrouk, M.; Miyazaki, H. Hydroxytyrosol induces proliferation and cytoprotection against oxidative injury in vascular endothelial cells: Role of Nrf2 activation and HO-1 induction. J. Agric. Food Chem. 2011, 59, 4473–4482. [Google Scholar] [CrossRef] [PubMed]
  14. Robles-Almazan, M.; Pulido-Moran, M.; Moreno-Fernandez, J.; Ramirez-Tortosa, C.; Rodriguez-Garcia, C.; Quiles, J.L.; Ramirez-Tortosa, M.C. Hydroxytyrosol: Bioavailability, toxicity, and clinical applications. Int. Food Res. J. 2018, 105, 654–667. [Google Scholar] [CrossRef] [PubMed]
  15. Li, D.; Zhang, W.; Tian, X.; He, Y.; Xiao, Z.; Zhao, X.; Fan, L.; Du, R.; Yang, G.; Yu, T. Hydroxytyrosol effectively improves the quality of pig sperm at 17 °C. Theriogenology 2022, 177, 172–182. [Google Scholar] [CrossRef] [PubMed]
  16. Branciari, R.; Galarini, R.; Giusepponi, D.; Trabalza-Marinucci, M.; Forte, C.; Roila, R.; Miraglia, D.; Servili, M.; Acuti, G.; Valiani, A. Oxidative status and presence of bioactive compounds in meat from chickens fed polyphenols extracted from olive oil industry waste. Sustainability 2017, 9, 1566. [Google Scholar] [CrossRef]
  17. Sabino, M.; Cappelli, K.; Capomaccio, S.; Pascucci, L.; Biasato, I.; Verini-Supplizi, A.; Valiani, A.; Trabalza-Marinucci, M. Dietary supplementation with olive mill wastewaters induces modifications on chicken jejunum epithelial cell transcriptome and modulates jejunum morphology. BMC Genom. 2018, 19, 576. [Google Scholar] [CrossRef]
  18. Pappas, A.C.; Tsiplakou, E.; Papadomichelakis, G.; Mitsiopoulou, C.; Sotirakoglou, K.; Mpekelis, V.; Haroutounian, S.A.; Fegeros, K.; Zervas, G. Effects of olive pulp addition to broiler diets on performance, selected biochemical parameters and antioxidant enzymes. J. Hellenic Vet. Med. Soc. 2019, 70, 1687–1696. [Google Scholar] [CrossRef]
  19. Papadomichelakis, G.; Pappas, A.C.; Tsiplakou, E.; Symeon, G.K.; Sotirakoglou, K.; Mpekelis, V.; Fegeros, K.; Zervas, G. Effects of dietary dried olive pulp inclusion on growth performance and meat quality of broiler chickens. Livest. Sci. 2019, 221, 115–122. [Google Scholar] [CrossRef]
  20. Dias, K.M.M.; Oliveira, C.H.; Calderano, A.A.; Rostagno, H.S.; O’Connor, K.E.; Davis, R.; Walsh, M.; Britton, J.; Altieri, E.A.; Albino, L.F.T. Effects of hydroxytyrosol supplementation on performance, meat quality and blood parameters of broiler chickens. Animals 2023, 14, 119. [Google Scholar] [CrossRef]
  21. Rostagno, H.S.; Albino, L.F.T.; Hannas, M.I.; Donzele, J.L.; Sakomura, N.K.; Perazzo, F.G.; Saraiva, A.; de Abreu, M.L.T.; Rodrigues, P.B.; de Oliveira, R.F.; et al. Brazilian Tables for Poultry and Swine: Composition of Feedstuffs and Nutritional Requirements, 4th ed.; Federal University of Viçosa: Viçosa, Brazil, 2017; 482p. [Google Scholar]
  22. Gava, M.S. Metodologia de Morfometria Intestinal em Frango de Corte. Master’s Thesis, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil, 2012. [Google Scholar]
  23. Luna, G.L. Manual of Histologic Staining Methods of the Armed Forces Institute of Pathology, 3rd ed.; Mc Graw-Hill Book Company: London, UK, 1968. [Google Scholar]
  24. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCt method. Methods 2011, 25, 402–408. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, W.; Li, Z.; Ren, W.; Yue, Y.; Guo, Y. Effects of live yeast supplementation on lipopolysaccharide-induced inflammatory responses in broilers. Poult. Sci. 2016, 95, 2557–2564. [Google Scholar] [CrossRef]
  26. Zhu, C.; Nie, X.; He, Z.; Xiong, T.; Li, Y.; Bai, Y.; Zhang, H. Research Note: Dietary resveratrol supplementation improves the hepatic antioxidant capacity and attenuates lipopolysaccharide-induced inflammation in yellow-feathered broilers. Poult. Sci. 2023, 102, 102370. [Google Scholar] [CrossRef] [PubMed]
  27. Jiang, J.; Qi, L.; Wei, Q.; Shi, F. Maternal stevioside supplementation ameliorates intestinal mucosal damage and modulates gut microbiota in the chick offspring challenged with lipopolysaccharide. Food Funct. 2021, 12, 6014–6028. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, Y.; Cheng, Y.; Wang, W.; Wang, A.; Zhou, Y. Protective effects of dietary supplementation with a silicate clay mineral (palygorskite) in lipopolysaccharide-challenged broiler chickens at an early age. Anim. Feed. Sci. Techonol. 2020, 263, 114459. [Google Scholar] [CrossRef]
  29. Lin, J.-Y.; Lai, Y.-S.; Liu, C.-J.; Wu, A.-R. Effects of lotus plumule supplementation before and following systemic administration of lipopolysaccharide on the splenocyte responses of BALB/c mice. Food Chem. Toxicol. 2007, 45, 486–493. [Google Scholar] [CrossRef] [PubMed]
  30. Yang, L.; Liu, G.; Zhu, X.; Luo, Y.; Shang, Y.; Gu, X.-L. The anti-inflammatory and antioxidant effects of leonurine hydrochloride after lipopolysaccharide challenge in broiler chicks. Poult. Sci. 2019, 98, 1648–1657. [Google Scholar] [CrossRef]
  31. Nunes, R.A.; Duarte, M.S.; Campos, P.H.R.F.; Oliveira, L.L.; Silva, F.F.; Kreuz, B.S.; Mirabile, C.G.; Borges, S.O.; Calderano, A.A. Active vitamin D3-glycoside preserves weight gain and modulates the inflammatory response in broiler chickens challenged with lipopolysaccharide. Anim. Feed Sci. Technol. 2020, 270, 114704. [Google Scholar] [CrossRef]
  32. Jiang, J.; Qi, L.; Lv, Z.; Jin, S.; Wei, X.; Shi, F. Dietary stevioside supplementation alleviates lipopolysaccharide-induced intestinal mucosal damage through anti-inflammatory and antioxidant effects in broiler chickens. Antioxidants 2019, 8, 575. [Google Scholar] [CrossRef]
  33. Zhang, Q.; Chen, X.; Eicher, S.D.; Ajuwon, K.M.; Applegate, T.J. Effect of threonine on secretory immune system using a chicken intestinal ex vivo model with lipopolysaccharide challenge. Poult. Sci. 2017, 96, 3043–3051. [Google Scholar] [CrossRef]
  34. Chang, Y.; Yuan, L.; Liu, J.; Muhammad, I.; Cao, C.; Shi, C.; Zhang, Y.; Li, R.; Li, C.; Liu, F. Dihydromyricetin attenuates Escherichia coli lipopolysaccharide-induced ileum injury in chickens by inhibiting NLRP3 inflammasome and TLR4/NF-κB signalling pathway. Vet. Res. 2020, 51, 72. [Google Scholar] [CrossRef]
  35. Liu, Y.; Huang, J.; Hou, Y.; Zhu, H.; Zhao, S.; Ding, B.; Yin, Y.; Yi, G.; Shi, J.; Fan, W. Dietary arginine supplementation alleviates intestinal mucosal disruption induced by Escherichia coli lipopolysaccharide in weaned pigs. Br. J. Nutr. 2008, 100, 552–560. [Google Scholar] [CrossRef] [PubMed]
  36. Li, Y.; Zhang, H.; Chen, Y.P.; Yang, M.X.; Zhang, L.L.; Lu, Z.X.; Zhou, Y.M.; Wang, T. Bacillus amyloliquefaciens supplementation alleviates immunological stress and intestinal damage in lipopolysaccharide-challenged broilers. Anim. Feed Sci. Technol. 2015, 208, 119–131. [Google Scholar] [CrossRef]
  37. Gao, J.; Zhang, H.J.; Yu, S.H.; Wu, S.G.; Yoon, I.; Quigley, J.; Gao, Y.P.; Qi, G.H. Effects of yeast culture in broiler diets on performance and immunomodulatory functions. Poult. Sci. 2008, 87, 1377–1384. [Google Scholar] [CrossRef] [PubMed]
  38. Montagne, L.; Pluske, J.R.; Hampson, D.J. A review of interactions between dietary fiber and the intestinal mucosa, and their consequences on digestive health in young non-ruminant animals. Anim. Feed Sci. Technol. 2003, 108, 95–117. [Google Scholar] [CrossRef]
  39. Yang, L.; Liu, G.; Lian, K.; Qiao, Y.; Zhang, B.; Zhu, X.; Luo, Y.; Shang, Y.; Gu, X.-L. Dietary leonurine hydrochloride supplementation attenuates lipopolysaccharide challenge-induced intestinal inflammation and barrier dysfunction by inhibiting the NF-κB/MAPK signaling pathway in broilers. J. Anim. Sci. 2019, 97, 1679–1692. [Google Scholar] [CrossRef]
  40. Wang, M.-Y.; Zhang, Y.; Tong, Y.-X.; Guo, P.-T.; Zhang, J.; Wang, C.-K.; Gao, Y.-Y. Effects of lutein on jejunal mucosal barrier function and inflammatory responses in lipopolysaccharide-challenged yellow-feather broilers. Poult. Sci. 2022, 101, 102191. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, W.; Zhou, J.; Chen, M.; Huang, X.; Xie, X.; Li, W.; Cao, Q.; Kan, H.; Xu, Y.; Ying, Z. Exposure to concentrated ambient PM2.5 alters the composition of gut microbiota in a murine model. Part. Fibre Toxicol. 2018, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  42. Keestra, A.M.; de Zoete, M.R.; Bouwman, L.I.; Vaezirad, M.M.; van Putten, J.P.M. Unique features of chicken Toll-like receptors. Dev. Comp. Immunol. 2013, 41, 316–323. [Google Scholar] [CrossRef]
  43. Li, W.; Guo, F.; Jiang, X.; Li, Y.; Li, X.; Yu, Z. Compound ammonium glycyrrhizin protects hepatocytes from injury induced by lipopolysaccharide/florfenicol through oxidative stress and a MAPK pathway. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2019, 225, 108585. [Google Scholar]
  44. Liu, S.D.; Song, M.H.; Yun, W.; Lee, J.H.; Kim, H.B.; Cho, J.H. Effect of carvacrol essential oils on immune response and inflammation-related genes expression in broilers challenged by lipopolysaccharide. Poult. Sci. 2019, 98, 2026–2033. [Google Scholar] [CrossRef]
  45. French, C.E.; Sales, M.A.; Rochell, S.J.; Rodriguez, A.; Erf, G.F. Local and systemic inflammatory responses to lipopolysaccharide in broilers: New insights using a two-window approach. Poult. Sci. 2022, 99, 6593–6605. [Google Scholar] [CrossRef] [PubMed]
  46. Arendt, M.; Elissa, J.; Schimdt, N.; Michael, E.; Potter, N.; Cook, M.; Knoll, L. Investigating the role of interleukin 10 on Eimeria intestinal pathogenesis in broiler chickens. Vet. Immunol. 2019, 218, 109934. [Google Scholar] [CrossRef]
  47. Kaiser, M.G.; Block, S.S.; Ciraci, C.; Fang, W.; Sifri, M.; Lamont, S.J. Effects of dietary vitamin E type and level on lipopolysaccharide-induced cytokine mRNA expression in broiler chicks. Poult. Sci. 2012, 91, 1893–1898. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, X.; Zhong, X.; Zhou, Y.; Wang, G.; Du, H.; Wang, T. Dietary RRR-a-tocopherol succinate attenuates lipopolysaccharide-induced inflammatory cytokines secretion in broiler chicks. Br. J. Nutr. 2010, 104, 1796–1805. [Google Scholar] [CrossRef] [PubMed]
  49. Zheng, X.C.; Wu, Q.J.; Song, Z.H.; Zhang, H.; Zhang, J.F.; Zhang, L.L.; Zhang, T.Y.; Wang, C.; Wang, T. Effects of Oridonin on growth performance and oxidative stress in broilers challenged with lipopolysaccharide. Poult. Sci. 2016, 95, 2281–2289. [Google Scholar] [CrossRef]
  50. Fukai, T.; Ushio-Fukai, M. Superoxide dismutases: Role in redox signaling, vascular function, and diseases. Antioxid. Redox Signal. 2011, 15, 1583–1606. [Google Scholar] [CrossRef] [PubMed]
  51. Satta, S.; Mahmoud, A.M.; Wilkinson, F.L.; Yvonne Alexander, M.; White, S.J. The role of Nrf2 in cardiovascular function and disease. Oxid. Med. Cell. Longev. 2017, 2017, 9237263. [Google Scholar] [CrossRef] [PubMed]
  52. Althunibat, O.Y.; Abduh, M.S.; Abukhalil, M.H.; Aladaileh, S.H.; Haineh, H.; Mahmoud, A.M. Umbelliferone prevents isoproterenol-induced myocardial injury by upregulating Nrf2/HO-1 signaling, and attenuating oxidative stress, inflammation, and cell death in rats. Biomed. Pharmacother. 2022, 149, 112900. [Google Scholar] [CrossRef]
  53. Wei, H.; Li, T.; Zhang, Y.; Liu, X.; Gong, R.; Bao, J.; Li, J. Cold stimulation causes oxidative stress, inflammatory response and apoptosis in broiler heart via regulating Nrf2/HO-1 and NF-κB pathway. J. Therm. Biol. 2023, 116, 103658. [Google Scholar] [CrossRef]
Figure 1. Effects of hydroxytyrosol supplementation on mRNA expression in the jejunum of broilers at 20 days old: (A) CAT, p value = 0.03; (B) GPx, p value = 0.0004; (C) IL-10, p value < 0.0001; (D) IL–1β, p value = 0.022; (E) IL–8, p value = 0.213; (F) NF–κB, p value = 0.633; and (G) SOD, p value = 0.315. Bars with different letters, along with their corresponding standard errors of the mean, differ from each other by the Tukey test. CON: control diet; CON + LPS: CON with LPS administration; HT + LPS: CON supplemented with 10 mg of hydroxytyrosol/kg of feed with LPS administration. CAT: catalase; GPx: glutathione peroxidase; IL–10: interleukin 10; IL–1β: interleukin 1β; IL–8: interleukin 8; NF–κB: nuclear factor kappa B; SOD: superoxide dismutase.
Figure 1. Effects of hydroxytyrosol supplementation on mRNA expression in the jejunum of broilers at 20 days old: (A) CAT, p value = 0.03; (B) GPx, p value = 0.0004; (C) IL-10, p value < 0.0001; (D) IL–1β, p value = 0.022; (E) IL–8, p value = 0.213; (F) NF–κB, p value = 0.633; and (G) SOD, p value = 0.315. Bars with different letters, along with their corresponding standard errors of the mean, differ from each other by the Tukey test. CON: control diet; CON + LPS: CON with LPS administration; HT + LPS: CON supplemented with 10 mg of hydroxytyrosol/kg of feed with LPS administration. CAT: catalase; GPx: glutathione peroxidase; IL–10: interleukin 10; IL–1β: interleukin 1β; IL–8: interleukin 8; NF–κB: nuclear factor kappa B; SOD: superoxide dismutase.
Animals 14 00871 g001
Table 1. Components and nutritional calculation of the basal diets (as-fed basis).
Table 1. Components and nutritional calculation of the basal diets (as-fed basis).
Ingredient8 to 20 Days, %
Corn, 7.88%50.38
Soybean meal, 45.0%41.12
Soybean oil4.58
Dicalcium phosphate1.67
Limestone0.84
Salt0.52
DL-Methionine, 99%0.32
L-Lysine, 78%0.15
L-Threonine, 98.5%0.05
Choline chloride, 60%0.10
Phytase 0.01
Antioxidant (BHT) 30.01
Mineral Premix 10.13
Vitamin Premix 20.13
Calculated nutritional composition 4
Crude Protein, %22.3
Metabolizable Energy, kcal/kg3000
Calcium, %0.980
Available Phosphorus, %0.480
Sodium, %0.220
Digestible Lysine, %1.240
Digestible Methionine, % 0.470
Digestible Methionine + Cysteine, % 0.929
Digestible Threonine, %0.810
Digestible Tryptophan, %0.267
1 Composition per kg of product: Manganese, 58.36 g; Iron, 41.68 g; Zinc, 54.21 g; Copper, 8.31 g; Iodine, 0.84 g; Selenium, 0.25 g. 2 Composition per kg of product: Vitamin A, 9,638,000 IU; Vitamin D3, 2,410,000 IU; Vitamin E, 36,100 IU; Vitamin B1, 2.60 g; Vitamin B2, 6.45 g; Vitamin B6, 3.61 g; Vitamin B12, 15.9 mg; Vitamin K3, 1.94 g; Pantothenic Acid, 12.95 g; Nicotinic Acid, 39.20 g; Folic Acid, 0.90 g; Biotin, 89.80 mg. 3 Antioxidant Butylhydroxytoluene. 4 Calculated following Rostagno et al. [21].
Table 2. Sequences of primers utilized in gene expression analysis.
Table 2. Sequences of primers utilized in gene expression analysis.
GeneGenBankSequence
NF-κBNM_205129.1F: 5′-GTGTGAAGAAACGGGAACTG-3′
R: 5′-GGCACGCTTGTCATAGATGG-3′
CATNM_001031215.2F: 5′-ACTGCAAGGCGAAAGTGTTT-3′
R: 5′-GGCTATGGATGAAGGATGGA-3′
GPXNM_001277853.2F: 5′-GACCAACCCGCAGTACATCA-3′
R: 5′-GAGGTGCGGGCTTTCCTTTA-3′
SOD1NM_205064.1F: 5′-AGGGGGTCATCCACTTCC-3′
R: 5′-CCCATTTGTGTTGTCTCCAA-3′
IL-10NM_001004414.2F: 5′-CATGCTGCTGGGCCTGAA-3′
R: 5′-CGTCTCCTTGATCTGCTTGATG-3′
IL-1βNM_204524.1F: 5′-GCTCTACATGTCGTGTGTGATGAG-3′
R: 5′-TGTCGATGTCCCGCATGA-3′
IL-8HM179639.1F: 5′-GGCTTGCTAGGGGAAATGA-3′
R: 5′-AGCTGACTCTGACTAGGAAACTGT-3′
β-ACTNM_205518.1F: 5′-TGCTGTGTTCCCATCTATCG-3′
R: 5′-TTGGTGACAATACCGTGTTCA-3′
Table 3. Feed intake (FI), body weight gain (BWG), body weight (BW), and feed conversion ratio (FCR) of broilers from 8 to 20 days.
Table 3. Feed intake (FI), body weight gain (BWG), body weight (BW), and feed conversion ratio (FCR) of broilers from 8 to 20 days.
CONCON + LPSHT + LPSp ValueSEM
FI, kg/bird0.9340.9260.9100.090.005
BWG, kg/bird0.741 a0.704 b0.736 a<0.0010.005
BW, kg/bird0.973 a0.932 b0.967 a<0.0010.005
FCR, kg/kg1.262 a1.317 b1.238 a<0.0010.010
a,b Letters differing within the same row indicate significant differences according to the Tukey Test (α = 0.05). SEM: standard error of the mean.
Table 4. Relative organ weight (ROW) of liver, spleen, and Bursa of broilers at 20 days old.
Table 4. Relative organ weight (ROW) of liver, spleen, and Bursa of broilers at 20 days old.
CONCON + LPSHT + LPSp ValueSEM
Liver, %2.494 b3.038 a2.853 a0.0010.067
Spleen, %0.099 b0.167 a0.150 a<0.00010.007
Bursa, %0.2140.2130.2130.9970.009
a,b Letters differing within the same row indicate significant differences according to the Tukey Test (α = 0.05). SEM: standard error of the mean.
Table 5. Villus height (VH), crypt depth (CD), and villus: crypt ratio (VH:CD) of jejunum from broiler chickens at 20 days old.
Table 5. Villus height (VH), crypt depth (CD), and villus: crypt ratio (VH:CD) of jejunum from broiler chickens at 20 days old.
CONCON + LPSHT + LPSp ValueSEM
VH, µm2100.851972.552108.570.32440.91
CD, µm477.77 a582.77 b448.81 a<0.00115.28
VH:CD4.453 a3.417 b4.879 a0.0040.198
a,b Letters differing within the same row indicate significant differences according to the Tukey Test (α = 0.05). SEM: standard error of the mean.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dias, K.M.M.; Oliveira, C.H.; Calderano, A.A.; Rostagno, H.S.; Gomes, K.M.; O’Connor, K.E.; Davis, R.; Walsh, M.; Britton, J.; Altieri, E.A.; et al. Dietary Hydroxytyrosol Supplementation on Growth Performance, Gut Morphometry, and Oxidative and Inflammatory Status in LPS-Challenged Broilers. Animals 2024, 14, 871. https://doi.org/10.3390/ani14060871

AMA Style

Dias KMM, Oliveira CH, Calderano AA, Rostagno HS, Gomes KM, O’Connor KE, Davis R, Walsh M, Britton J, Altieri EA, et al. Dietary Hydroxytyrosol Supplementation on Growth Performance, Gut Morphometry, and Oxidative and Inflammatory Status in LPS-Challenged Broilers. Animals. 2024; 14(6):871. https://doi.org/10.3390/ani14060871

Chicago/Turabian Style

Dias, Kelly M. M., Carlos H. Oliveira, Arele A. Calderano, Horacio S. Rostagno, Kaique M. Gomes, Kevin E. O’Connor, Reeta Davis, Meg Walsh, James Britton, Enrico A. Altieri, and et al. 2024. "Dietary Hydroxytyrosol Supplementation on Growth Performance, Gut Morphometry, and Oxidative and Inflammatory Status in LPS-Challenged Broilers" Animals 14, no. 6: 871. https://doi.org/10.3390/ani14060871

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop