Next Article in Journal
Computational Insights into Reproductive Toxicity: Clustering, Mechanism Analysis, and Predictive Models
Previous Article in Journal
Evaluation of SARS-CoV-2-Specific IgY Antibodies: Production, Reactivity, and Neutralizing Capability against Virus Variants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploring the Effects of an Alfalfa Leaf-Derived Adsorbent on Microbial Community, Ileal Morphology, Barrier Function, and Immunity in Turkey Poults during Chronic Aflatoxin B1 Exposure

by
María de Jesús Nava-Ramírez
1,
Jing Liu
2,*,
Juan Omar Hernández-Ramírez
1,
Xochitl Hernandez-Velasco
3,
Juan D. Latorre
4,
Alma Vázquez-Durán
1,
Guolong Zhang
2,
Roberto Senas-Cuesta
4,
Sergio Gómez-Rosales
5,
Andressa Stein
4,
Billy M. Hargis
4,
Guillermo Téllez-Isaías
4,
Abraham Méndez-Albores
1 and
Jesús A. Maguey-González
4,*
1
Unidad de Investigación Multidisciplinaria L14 (Alimentos, Micotoxinas, y Micotoxicosis), Facultad de Estudios Superiores (FES) Cuautitlán, UNAM, Cuautitlán Izcalli 54740, Mexico
2
Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
3
Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia, UNAM, Ciudad de México 04510, Mexico
4
Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
5
Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal (CENID-INIFAP), Km 1 Carretera a Colon, Ajuchitlán, Querétaro 76280, Mexico
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(14), 7977; https://doi.org/10.3390/ijms25147977 (registering DOI)
Submission received: 14 May 2024 / Revised: 12 July 2024 / Accepted: 15 July 2024 / Published: 22 July 2024
(This article belongs to the Section Molecular Microbiology)

Abstract

:
This article follows-up on our recently published work, which evaluated the impact of the addition of an alfalfa leaf-derived adsorbent in the aflatoxin B1 (AFB1)-contaminated diet in regard to the production parameters, blood cell count, serum biochemistry, liver enzymes, and liver histology of turkey poults. This paper presents complementary results on microbial community, ileal morphology, barrier function, and immunity. For this purpose, 350 1-day-old female turkey poults were randomly distributed into five groups: (1) Control, AFB1-free diet; (2) AF, AFB1-contaminated diet at 250 ng/g; (3) alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; (4) alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and (5) YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group). In general, in the AF group, the growth of opportunistic pathogens was promoted, which lead to gut dysbacteriosis, mainly influenced by Streptococcus lutetiensis. Conversely, a significant increase in beneficial bacteria (Faecalibacterium and Coprococcus catus) was promoted by the addition of the plant-based adsorbent. Moreover, the AF group had the lowest villus height and a compromised barrier function, as evidenced by a significant (p < 0.05) increase in fluorescein isothiocyanate dextran (FITC-d), but these negative effects were almost reversed by the addition of the alfalfa adsorbent. Furthermore, the AF + YCW and alfalfa + AF groups exhibited a significant increase in the cutaneous basophil hypersensitivity response compared to the rest of the experimental groups. Taken together, these results pointed out that the alfalfa counteracts the adverse effects of AFB1 in poults, facilitating the colonization of beneficial bacteria and improving the barrier function of the turkey poults.

1. Introduction

Aflatoxin B1 (AFB1) is a potent secondary metabolite mainly synthesised by Aspergillus spp. AFB1 contamination in poultry feed negatively influences gut health, immunity, and performance. As a result, AFB1 contamination has become a major preoccupation in the world of poultry producers and researchers [1]. Consequently, to avoid these adverse effects, multiple strategies have been developed to control AFB1 in feed and to improve the health of broiler birds in the poultry industry. Recently, functional natural feed additives have been considered as promising substitutes for antibiotic growth promoters due to their antioxidant, anti-inflammatory, and immune-modulation properties [2].
While maize and soy remain the foundation of many (if not most) broiler diets, supplementation with nutrient-dense forages such as alfalfa could offer several advantages. Packed with vitamins, minerals, and bioactive compounds, alfalfa stands to enhance the environmental profile of broiler production while promoting the health and welfare of the chicken itself. First, alfalfa is nutritious, its protein content is high, and, as such, it is an excellent building block for muscle. Moreover, vitamins A, E, and K, along with its calcium, magnesium, and iron content, provide health support and promote strong and healthy bones [3]. Alfalfa’s carotene content also contributes to the yellow colouration of the chicken skin and yolk, something that is linked to important economic preference in certain consumer markets [4]. Concerning gut health, alfalfa’s high fibre content stimulates the production of beneficial gut bacteria, helping to keep the chicken healthy and enhance their efficiency [5]. In this context, some researchers have also suggested the possibility of having higher meat quality when alfalfa is included in broiler diets [6]. Moreover, the high level of natural antioxidants contributes to increasing the shelf life of the meat as well as reducing lipid oxidation [7].
Feeding alfalfa also brings about environmental benefits. The nitrogen-fixing ability of alfalfa reduces fertiliser application in a pasture, while its deep roots improve soil health and the retention of water [8]. These benefits can be translated to lower environmental impact and higher sustainability during broiler production.
The outcomes from the recent in vitro [9] and in vivo [10] studies from our laboratories confirmed the efficacy of alfalfa in ameliorating AFB1 toxicity and enhancing the performance in turkey poults. The present study continues the same experimental design to determine the effects of alfalfa on the composition and diversity of the gut microbial community, intestinal permeability, the morphometric analysis of ileum, and the cellular immunity. This research complements and indicates crucial roles of alfalfa leaves in the protection against aflatoxin B1-induced toxicity, suggesting that this intervention can be used as a novel approach to improve intestinal health and enhance resilience in turkey poults during aflatoxicosis.

2. Results

There were no obvious distinctions among the experimental groups in terms of α-diversity (Figure 1). Nevertheless, the AF group exhibited a lower diversity (number of observed ASVs, Evenness, and Shannon indexes) than the Control group, indicating that the challenge to AFB1 could potentially result in the proliferation of specific pathogenic bacteria, which could contribute to a change in the bacterial community. However, supplementation of alfalfa could almost restore this decreased trend of bacteria to healthy levels (Figure 1).
Similarly, there was no significant separation among groups based on the weighted UniFrac distance (Figure 1). However, an unweighted UniFrac distance showed a significant difference among the Control and AF groups, indicating the influence of AFB1 on the microbial community composition (p < 0.05). In addition, a clear separation between the AF group and the AF + YCW group was also observed (p = 0.017, R2 = 0.168), suggesting that YCW supplementation during the AFB1 challenge could also change the bacterial community structure (β-diversity).
Table 1 summarises the findings of the relative abundances (%) of the cecal bacterial phyla and their families between the experimental groups. The contamination with 250 ng AFB1/g did not affect the phyla relative abundances of the microbial community in the caecum of any of the experimental groups. However, at the family level, a significant decrease in Streptococcaceae populations was observed in the Control group compared to the rest of the experimental groups (p < 0.04). In addition, the relative genera abundance of Coprobacillaceae was reduced in the AF and alfalfa + AF groups, and the same pattern was observed in the ASV levels (p < 0.02). On the other hand, the alfalfa group exhibited a substantial increase in Faecalibacterium levels in contrast to the other experimental groups (Table 2). At the ASV level, bacterial biomarkers for each group were identified using linear discriminant analysis effect size (LEfSe), employing an all-against-all multiclass analysis approach (Figure 2). Several bacterial species were found to be enriched in the LEfSe analysis using the data from all experimental groups. For instance, the signature ASVs for the Control group were Mediterraneibaer (F91) and Lachnospiracaeae unidentified (F25), while AF group had greater abundances of Streptococcus lutetiensis (F6). In addition, the alfalfa group were Faecalibacterium (F7 and F53) and Coprococcus catus (F85), while the alfalfa + AF group had greater abundances of Bacillus (F8) and Anaerotignum (F99).
In addition, Figure 3 shows the relative abundances of differentially enriched bacterial ASVs. Alfalfa administration led to a substantial increase in Faecalibacterium and C. catus (p < 0.05). Moreover, the populations of Bacillus and Anaerotignum, as well as microorganisms related to beneficial bacteria populations and producers of short-chain fatty acids (SCFAs), exhibited a substantial increase in poults that were fed with alfalfa and challenged with AFB1 (p < 0.05). On the other hand, the results of the morphometric analysis, serum levels of FITC-d, and cutaneous basophil hypersensitivity response (CBH) are summarised in Table 3. In general, the AF group had the lowest villus height (p < 0.0001) and the total villus cross-sectional area (p < 0.0001). Meanwhile, the Control (780.0 μm), AF + YCW (700.0 μm), alfalfa (693.3 μm), and alfalfa + AF (611.9 μm) groups showed a significant increase in villus height, followed by the AF (350.6 μm) group. Similarly, a significant increase in the total villus cross-sectional area was observed in the Control (103.2 μm2), AF + YCW (74.1 μm2), alfalfa (71.7 μm2), and alfalfa + AF (63.6 μm2) groups, followed by the AF (44.7 μm2) group (Figure 4). Furthermore, the AF group had the greatest serum FITC-d concentration (p < 0.007) in comparison to the other experimental groups. Moreover, poults of the AF + YCW (0.96 mm) and alfalfa + AF (0.86 mm) groups showed a significant increase in CBH response (p < 0.0001) in comparison to the rest of the experimental groups.

3. Discussion

In this work, the inclusion of powdered alfalfa leaves into the AFB1-contaminated feed confirmed the adsorbent properties of this material previously described in in vitro [9] and in vivo [10] assays. AFB1-contaminated feed may lower the growth rate and feed efficiency of poultry. However, due to the high protein content and balanced mineral composition, alfalfa may partly alleviate these adverse effects by supporting optimal growth for turkey poults. Thus, the adsorbent properties of alfalfa against AFB1 explain the reason for having improved growth of poults challenged with AFB1 [10]. Furthermore, alfalfa provides vitamins, minerals, fibre, and proteins that enhance chickens’ diets and favour wellness and overall performance. Notably, the natural antioxidants present in alfalfa, such as vitamins A, C, and E, help to prevent the oxidative stress induced by AFB1 [11]. Indeed, AFB1 could generate a series of reactive oxygen species, and antioxidants in alfalfa might increase the ability of broilers to counteract oxidative damage. In addition, alfalfa contains a diverse array of immune-boosting compounds [12]. By boosting the immune system, alfalfa helps broiler chickens manage the immunosuppressive effects of AFB1 [13]. It is well known that AFB1 primarily attacks the liver when administered to poultry [14], causing increased liver damage while lowering the antioxidant status in the liver tissue and plasma. Alfalfa’s potential detoxification properties and nutrient profile may prove useful in maintaining liver function and preventing the damage of the liver from toxins such as AFB1 [15].
In the present study, the LEfSE analysis likely identified S. lutetiensis as the most abundant bacterial species in the AFB1-treated group. This finding suggests that the presence of AFB1 improves the proliferation of S. lutetiensis in the gut of turkey poults. The toxic effects of AFB1 can affect both cell-mediated and humoral immunity in poultry, thus facilitating the entry and invasion of bacterial pathogens [16]. Hence, the mycotoxin-induced immunosuppression promotes the colonization of opportunistic pathogens like S. lutetiensis in the gut of turkey poults [17,18]. In addition, AFB1 has been reported to cause dysbacteriosis [19] and impair the nutrient absorption capacity, weaken their immune system, and generate a more suitable environment for S. lutetiensis growth [20].
On the other hand, the LEfSE analysis also showed an enhanced abundance of Faecalibacterium within the alfalfa-fed group compared with the positive control group (poults that received the AFB1), possibly due to the intricate interactions between alfalfa’s properties and AFB1’s toxicity. Alfalfa has plenty of bioactive compounds, such as saponins and phenolic acids, which have the potential to bind AFB1 within the gut, thus reducing its absorption and minimising its toxicity [21]. This finding suggests that Faecalibacterium, rather than being inhibited by the AFB1 challenge, has flourished within a friendlier gut environment. Furthermore, alfalfa is replete with dietary fibres, particularly oligosaccharides and fructooligosaccharides (FOS), acting as prebiotics—substances that stimulates the growth and activity of a specific, beneficial gut bacteria—such as Faecalibacterium [22]. FOS have also been reported to exhibit antimicrobial activity by prohibiting pathogenic microbes from adhering to the intestinal epithelium. Furthermore, alfalfa demonstrates antioxidant potency, most notably by carotenoids and phenolic acids [23]. These antioxidants battle the oxidative stress that stems from AFB1 and, consequently, promote friendlier gut environments for Faecalibacterium growth [21]. Interestingly, a recent study has demonstrated a natural resilience of Faecalibacterium towards AFB1 [24] while also documenting the production of anti-inflammatory metabolites, especially butyrate, which can help ameliorate the gut inflammation linked to AFB1 [25]. The combination of Faecalibacterium proliferation with those anti-inflammatory metabolites may in turn facilitate the proliferation of other health-promoting microbes and limiting pathogenic microbes by reducing their growth and their ability to colonise the gastrointestinal tract [26]. Taken together, the competitive microbial advantage, in combination with the changes to the gut environment through alfalfa supplementation, could add to the explanation for the Faecalibacterium’s increased abundance.
Another notation worthy of mentioning by the LEfSE analysis is the increased relative abundance of C. catus in turkey poults that received alfalfa and AFB1. C. catus belongs to the class Clostridia, order Eubacteriales. Recently, investigators have noticed favourable correlations between C. catus and health [27]. Consequently, it is plausible to speculate that the increased abundance in C. catus in the turkey poults that received the diet contaminated with AFB1 might be attributed to the capability of the bacterium to resist and metabolise AFB1 [28]. This in turn will be a selective advantage for it over other bacteria, which succumb to the toxic effects of AFB1 [29]. Interestingly, exposure of prokaryotes and eukaryotes to AFB1 has been documented as a cause of stress [30]. C. catus has been shown to exhibit enhanced stress tolerance in comparison with other gut microbiota [31], which might afford it superior survival in the gut stressed by AFB1 and ultimately increase its relative abundance.
The notable reduction in the serum concentration of FITC-d in turkey poults fed with alfalfa and AFB1 compared with the positive control group may be influenced by several factors. For instance, AFB1 can induce intestinal inflammation and increase intestinal permeability [32]. On the other hand, alfalfa is rich in bioactive compounds such as tannins and phenolic acids, which are well known for their binding properties against AFB1 in the digestive tract, particularly in the intestine, reducing its absorption [33]. Consequentially, this should result in a lower serum concentration of FITC-d. Moreover, alfalfa is rich in antioxidants, mainly expressed by the presence of chlorophyll, carotenoids, and vitamin E ([34]. These bioactive compounds have the capability of scavenging free radicals generated during the AFB1 metabolism and, ultimately, reducing the degree of oxidative damage, which could be responsible for the translocation of FITC-d, by affecting intestinal epithelial cells [34]. Overall, a better intestinal barrier will contribute to a better intestinal integrity. Furthermore, alfalfa can also improve the performance of the gastrointestinal tract by augmenting populations of beneficial bacteria and suppressing those considered harmful [6]. The shift from ‘harmful’ to ‘beneficial’ gut microbiota will be essential in increasing the efficiency of bacteria responsible for AFB1 degradation, as well as for competing with the mycotoxin for binding sites in the intestine, all of which will ultimately decrease its bioavailability. Alternatively, the immunomodulatory action of alfalfa [35] and its anti-inflammatory properties [36] could contribute to the epithelial tissue repair in the intestinal epithelium and, therefore, reduce the FITC-d leakage.
In addition, the increase in ileum villus height in the AFB1-treated turkey poults with the addition of alfalfa as compared with the Control birds is quite interesting, but the exact mechanism is complex. AFB1 is a very potent hepatotoxin; however, exposure to this mycotoxin can also impair functions of the intestine, which can result in damage to the lining of villus where the enterocytes are situated. This damage can result in a reduction in the capacity of the body to absorb nutrients and can also have an impact on the overall health of the gut. Despite the damage to the enterocytes lining the villus, the body might try to compensate for this damage by increasing the height of the villus in order to maintain a greater surface area for absorption. This effect could possibly be viewed as a mechanism to help the body adapt over the short-term exposure. Moreover, fibres and other bioactive compounds from alfalfa could be prebiotic to beneficial gut bacteria, thus promoting their growth and maintaining the health of the gut and the absorption of nutrients [37]. For instance, increasing beneficial bacteria and their metabolites could stimulate villus growth and improve intestinal function. The antioxidants present in alfalfa might also reduce oxidative stress because of AFB1, which, in turn, could be beneficial in regard to healthier villus [23].

4. Materials and Methods

4.1. Animal Source, Diets, and Experimental Design

Three hundred and fifty 1-day-old female Nicholas-700 turkey poults (Aviagen Inc., Huntsville, AR, USA) were raised in pens for 28 d. Yeast cell wall (YCW) was used as a reference material because it is a commercial mycotoxin binder. Poults were collectively weighted (10 birds/pen) with 7 repetitions per treatment and randomly allocated to one of the five experimental groups: (1) Control, AFB1-free diet; (2) AF (aflatoxin), AFB1-contaminated diet at 250 ng/g; (3) alfalfa (alfalfa adsorbent), AFB1-free diet + 0.5% (w/w) adsorbent; (4) alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and (5) YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent. The percentage of adsorbents added to the diets is equivalent to 5 g of adsorbent/kg of feed consumed. Details of the experimental diet contaminated with AFB1 and the adsorbent material are fully described in our previous study [9]. Water and feed were offered ad libitum. After 28 d, from each replication, three poults (n = 21) were selected to evaluate gut integrity and cellular immunity. One poult from each replication (n = 7) was also selected to assess the morphometry of the ileum and ceca content for the microbiota analysis. All animal handling procedures complied with the Institutional Animal Care and Use Committee (IACUC) at the University of Arkansas, Fayetteville (protocol No. 22020).

4.2. Microbial Community

Following euthanasia, one cecum was removed, and the contents were manually squeezed into a sterile tube featuring a RNA/DNA shield (Zymo Research, Irvine, CA, USA). Samples were stores at −20 °C until DNA extraction was performed. A Quick-DNA Fecal/Soil Mi-crobe Miniprep Kit (Zymo Research, Irvine, CA, USA) was used to extract microbial DNA from the ceca contents following the manufacturer’s instructions. With the NanoDrop ND-1000 (Wilmington, DE, USA), the DNA concentration and quality were assessed. The bacterial 16S rRNA gene’s V3–V4 region was amplified using primers 341F (CCTAYGGGRBGCASCAG) and 806R (GGACTAC-NNGGGTATCTAAT). A NEBNext® UltraTM Library Prep Kit (New England Biolabs, Ipswich, MA, USA) was used to produce a library, which was then sequenced using PE250 on an Illumina HiSeq platform.

4.3. Bioinformatics and Statistical Analysis

The QIIME 2 pipeline (v. 2023.07) was employed to analyse raw DNA sequencing reads. In summary, the cut–adapt plugin was used to eliminate primer and adapter sequences from every read. After that, low-quality reads were eliminated using the quality filter and paired-end reads were combined using VSEARCH join pairs. Sequences were then trimmed to 403 nucleotides and denoised by Deblur [38]. The resultant amplicon sequence variants (ASVs) were then classified using a Bayesian classifier and the Ribosomal Database Project (RDP) 16S rRNA training set (v. 18) for the bacterial taxonomy. For categorization, a bootstrap confidence of 80% was applied. ASVs with a classification of <80% were assigned the name of the last confidently assigned level followed by “unclassified”. The analysis excluded samples with ASVs found in less than 5% of the samples. The EzBioCloud 16S database (v.2023.08.23. https://www.ezbiocloud.net/identify Accessed on 28 November 2024) was used to further confirm Tophree 50 ASVs and other differentially enriched microorganisms, reclassifying them as needed.

4.4. Ileal Morphology

A hematoxylin and eosin (H&E) staining technique was routinely applied to ileum samples that were obtained midway between Meckel’s diverticulum and the ileocecal junction. The samples were cut into 5 μm thick sections and embedded in paraffin. Photomicrographs were taken using a camera ICC50W associated with a microscope Leica DM2500 (Leica, Wetzlar, Germany). The variables measured were the villus height, villus width, and the villus cross-sectional area (villus height × villus width). Morphometric measurements were performed using the ImageJ 1.52v software. Every variable in each treatment considered 63 measurements (9 per replicate).

4.5. Barrier Function

Intestinal permeability was assessed by measuring the levels of the biomarker fluorescein isothiocyanate dextran (FITC-d, molecular weight 3–5 kDa, Merck KGaA, Darmstadt, Germany) in the serum. To achieve this, FITC-d (8.32 mg/kg) was given orally to twenty-one poults per treatment one hour before euthanasia. The serum samples were processed according to the recommendations of Baxter et al. [39], and fluorescence measurements were performed at 485 nm excitation and 528 nm emission using a Synergy HT multimode micro plate reader (Bio Tek Instruments, Inc., Winooski, VT, USA).

4.6. Immunity

The skin response to phytohemagglutinin m (PHA-M) was used to measure cellular immune activity using cutaneous basophil hypersensitivity (CBH). On day 28, three poults per replicate were randomly selected and injected intradermally in the interdigital skin between the third and fourth digits of the left foot with 0.1 mL of PHA m (Gibco, Grand Island, NY, USA). The CBH response was calculated by using the following mathematical expression:
C B H   m m = t h i c k n e s s   24   h   p o s t i n j e c t i o n ( t h i c k n e s s   p r e i n j e c t i o n )

4.7. Statistical Analysis

The ASV tables were standardised through the application of cumulative sum scaling (CSS) within the R (v. 1.4.0) metagenome Seq package [40]. The phyloseq package v. 1.42.0 was used to calculate the α-diversity (Shannon’s Index, Observed ASV, and Pielou’s Evenness) and β-diversity (unweighted and weighted UniFrac distances), which were then plotted using the ggplot2 program in R [41]. The statistical significance of α-diversity and relative abundance was assessed by a non-parametric Kruskal–Wallis test, which was followed by a pairwise Wilcoxon rank sum test. Using the ADONIS function in the vegan package v. 2.6.4, the non-parametric permutational multivariate analysis of variance (PERMANOVA) was used to determine the significance in β-diversity [42]. To determine the differential enrichment of bacteria ASVs among different groups of bacteria, linear discriminant analysis (LDA) effect size (LEfSe) was employed, with a threshold of p < 0.05 and an LDA score of ≥3.0 [43]. Finally, morphometric, serum concentrations of FITC-d, and CBH data were all subjected to ANOVA in a completely randomised design, utilising the general linear model process in SAS [44]. By using the Tukey multiple range test at p < 0.05, means were separated.

4.8. Data Availability

Raw sequencing reads were deposited in the NCBI GenBank SRA database under the accession number PRJNA1020155.

5. Conclusions

In the present study, the results indicated that the addition of alfalfa adsorbent promoted the proliferation of beneficial bacteria, like Faecalibacterium and Coprococcus catus, increased the height of the villi and significantly enhanced intestinal permeability, which was in contrast to the AF group. Finally, we evaluated the complex link between dietary supplementation with alfalfa, AFB1, and the cellular immune response. The significant increase in CBH in response to phytohemagglutinin injection in turkey poults supplemented with alfalfa and AFB1 may be explained by the fact that alfalfa and AFB1 interact to boost the immunological response, since alfalfa contains components that may counteract the immunosuppressive effects of AFB1, resulting in a more robust immune response. In conclusion, the results of this study indicate that alfalfa may serve as an effective dietary intervention for the mitigation of the adverse effects of AFB1 in turkey poults.

Author Contributions

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

Funding

This project was funded by UNAM-PAPIIT grant number IA101523 and by funds provided by USDA Animal Health Awards (FY2021 & FY2022), as well as by USDA-NIFA Sustainable Agriculture Systems, Grant No. 2019-69012-29905. Title of Project: Empowering U.S. Broiler Production for Transformation and Sustainability USDA-NIFA (Sustainable Agriculture Systems): No. 2019-69012-29905.

Institutional Review Board Statement

All animal handling procedures complied with the Institutional Animal Care and Use Committee (IACUC) at the University of Arkansas, Fayetteville (protocol No. 22020).

Informed Consent Statement

Not applicable.

Data Availability Statement

Upon reasonable request, and subject to review, the authors will provide the data that support the findings of this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ghaemmaghami, S.S.; Rouhanipour, H.; Sharifi, S.D. Aflatoxin levels in poultry feed: A comparison of mash and pellet forms. Poult. Sci. 2024, 103, 103254. [Google Scholar] [CrossRef] [PubMed]
  2. Fathima, S.; Al Hakeem, W.G.; Selvaraj, R.K.; Shanmugasundaram, R. Beyond protein synthesis: The emerging role of arginine in poultry nutrition and host-microbe interactions. Front. Physiol. 2024, 14, 1326809. [Google Scholar] [CrossRef] [PubMed]
  3. Kim, W.K.; Herfel, T.M.; Dunkley, C.S.; Hester, P.Y.; Crenshaw, T.D.; Ricke, S.C. The effects of alfalfa-based molt diets on skeletal integrity of white leghorns. Poult. Sci. 2008, 87, 2178–2185. [Google Scholar] [CrossRef] [PubMed]
  4. Yıldız, A.Ö.; Şentürk, E.T.; Olgun, O. Use of alfalfa meal in layer diets–a review. J. World’s Poult. Sci. 2020, 76, 134–143. [Google Scholar] [CrossRef]
  5. Jha, R.; Mishra, P. Dietary fiber in poultry nutrition and their effects on nutrient utilization, performance, gut health, and on the environment: A review. J. Anim. Sci. Biotechnol. 2021, 12, 51. [Google Scholar] [CrossRef]
  6. Chen, X.; Li, X.; Chen, D.; Zhao, W.; Zhang, X.; Yuan, W.; Si, H.; Deng, X.; Du, R.; Xu, C. Effects of dietary alfalfa meal supplementation on the growth performance, nutrient apparent digestibility, serum parameters, and intestinal microbiota of raccoon dogs (Nyctereutes procyonoides). Animals 2024, 14, 623. [Google Scholar] [CrossRef] [PubMed]
  7. Ouyang, K.; Xu, M.; Jiang, Y.; Wang, W. Effects of alfalfa flavonoids on broiler performance, meat quality, and gene expression. Can. J. Anim. Sci. 2021, 96, 332–341. [Google Scholar] [CrossRef]
  8. Qin, P.; Hu, L.; Liu, Y.; Hu, X.; Zhang, X.; Rosado, A.S.; Wei, G.; Chen, C. Responses of soil microbial communities and nutrient dynamics under continuous alfalfa (Medicago sativa L.) cultivation. Appl. Soil Ecol. 2024, 197, 105356. [Google Scholar] [CrossRef]
  9. Nava-Ramírez, M.J.; Vázquez-Durán, A.; Figueroa-Cárdenas, J.d.D.; Hernández-Patlán, D.; Solís-Cruz, B.; Téllez-Isaías, G.; López-Coello, C.; Méndez-Albores, A. Removal of aflatoxin B1 using alfalfa leaves as an adsorbent material: A comparison between two in vitro experimental models. Toxins 2023, 15, 604. [Google Scholar] [CrossRef] [PubMed]
  10. Nava-Ramírez, M.J.; Maguey-González, J.A.; Gómez-Rosales, S.; Hernández-Ramírez, J.O.; Latorre, J.D.; Du, X.; López-Coello, C.; Hargis, B.M.; Téllez-Isaías, G.; Vázquez-Durán, A.; et al. Efficacy of powdered alfalfa leaves to ameliorate the toxic effects of aflatoxin B1 in turkey poults. Mycotoxin Res. 2024, 40, 269–277. [Google Scholar] [CrossRef]
  11. Alpsoy, L.; Yalvac, M.E. Key roles of vitamins A, C, and E in aflatoxin B1-induced oxidative stress. Vitam. Horm. 2024, 2011, 287–305. [Google Scholar] [CrossRef]
  12. Ahmadi, A.; Shahidi, S.A.; Safari, R.; Motamedzadegan, A.; Ghorbani-HasanSaraei, A. Evaluation of stability and antibacterial properties of extracted chlorophyll from alfalfa (Medicago sativa L.). Food Chem. Toxicol. 2022, 163, 112980. [Google Scholar] [CrossRef] [PubMed]
  13. Mohsenzadeh, M.S.; Hedayati, N.; Riahi-Zanjani, B.; Karimi, G. Immunosuppression following dietary aflatoxin B1 exposure: A review of the existing evidence. Toxin Rev. 2016, 35, 121–127. [Google Scholar] [CrossRef]
  14. Quezada, T.; Cuellar, H.; Jaramillo-Juarez, F.; Valdivia, A.G.; Reyes, J.L. Effects of aflatoxin B1 on the liver and kidney of broiler chickens during development. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2000, 125, 265–272. [Google Scholar] [CrossRef] [PubMed]
  15. Christou, A.; Antoniou, C.; Christodoulou, C.; Hapeshi, E.; Stavrou, I.; Michael, C.; Fatta-Kassomps, D.; Fotopoulos, V. Stress-related phenomena and detoxification mechanisms induced by common pharmaceuticals in alfalfa (Medicago sativa L.) plants. Sci. Total Environ. 2016, 557, 652–664. [Google Scholar] [CrossRef] [PubMed]
  16. Ditta, Y.A.; Mahad, S.; Bacha, U. Aflatoxins: Their toxic effect on poultry and recent advances in their treatment. In Mycotoxins-Impact and Management Strategies; IntechOpen: London, UK, 2018. [Google Scholar] [CrossRef]
  17. Crispo, M.; Shivaprasad, H.L.; Cooper, G.L.; Bickford, A.A.; Stoute, S.T. Streptococcosis in commercial and noncommercial avian species in California: 95 cases (2000–2017). Avian Dis. 2018, 62, 152–162. [Google Scholar] [CrossRef] [PubMed]
  18. Maguey-González, J.A.; Liu, J.; Zhang, G.; Latorre, J.D.; Hernández-Ramírez, J.O.; de Jesús Nava-Ramírez, M.; Senas-Cuesta, R.; Gómez-Rosales, S.; de Lourdes Ángeles, M.; Stein, A.; et al. Assessment of the impact of humic acids on intestinal microbiota, gut integrity, ileum morphometry, and cellular immunity of turkey poults fed an aflatoxin B1-contaminated diet. Toxins 2024, 16, 122. [Google Scholar] [CrossRef]
  19. Huang, S.; Lin, L.; Wang, S.; Ding, W.; Zhang, C.; Shaukat, A.; Xu, B.; Yue, K.; Zhang, C.; Liu, F. Total flavonoids of Rhizoma drynariae mitigates aflatoxin B1-induced liver toxicity in chickens via microbiota-gut-liver axis interaction mechanisms. Antioxidants 2023, 12, 819. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, X.; Naehrer, K.; Applegate, T.J. Interactive effects of dietary protein concentration and aflatoxin B1 on performance, nutrient digestibility, and gut health in broiler chicks. Poult. Sci. 2016, 95, 1312–1325. [Google Scholar] [CrossRef]
  21. Wang, L.; Gao, M.; Kang, G.; Huang, H. The potential role of phytonutrients flavonoids influencing gut microbiota in the prophylaxis and treatment of inflammatory bowel disease. Front. Nutr. 2021, 8, 798038. [Google Scholar] [CrossRef] [PubMed]
  22. Kim, W.K.; Donalson, L.M.; Mitchell, A.D.; Kubena, L.F.; Nisbet, D.J.; Ricke, S.C. Effects of alfalfa and fructooligosaccharide on molting parameters and bone qualities using dual energy X-ray absorptiometry and conventional bone assays. Poult. Sci. 2006, 85, 15–20. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, X.; Guo, X.S.; Li, F.H.; Usman, S.; Zhang, Y.X.; Ding, Z.T. Antioxidant, flavonoid, α-tocopherol, β-carotene, fatty acids, and fermentation profiles of alfalfa silage inoculated with novel Lactiplantibacillus plantarum and Pediococcus acidilactici strains with high-antioxidant activity. Anim. Feed. Sci. Technol. 2022, 288, 115301. [Google Scholar] [CrossRef]
  24. Jin, J.; Beekmann, K.; Ringø, E.; Rietjens, I.M.; Xing, F. Interaction between food-borne mycotoxins and gut microbiota: A review. Food Control 2021, 126, 107998. [Google Scholar] [CrossRef]
  25. Sitkin, S.; Pokrotnieks, J. Clinical potential of anti-inflammatory effects of Faecalibacterium prausnitzii and butyrate in inflammatory bowel disease. Inflamm. Bowel Dis. 2019, 25, e40–e41. [Google Scholar] [CrossRef] [PubMed]
  26. Nyangale, E.P.; Farmer, S.; Cash, H.A.; Keller, D.; Chernoff, D.; Gibson, G.R. Bacillus coagulans GBI-30, 6086 modulates Faecalibacterium prausnitzii in older men and women. J. Nutr. 2015, 145, 1446–1452. [Google Scholar] [CrossRef] [PubMed]
  27. Yang, R.; Shan, S.; Shi, J.; Li, H.; An, N.; Li, S.; Ciu, K.; Guo, H.; Li, Z. Coprococcus eutactus, a potent probiotic, alleviates colitis via acetate-mediated IgA response and microbiota restoration. J. Agric. Food Chem. 2023, 71, 3273–3284. [Google Scholar] [CrossRef] [PubMed]
  28. Belanche, A.; Palma-Hidalgo, J.M.; Jiménez, E.; Yáñez-Ruiz, D.R. Enhancing rumen microbial diversity and its impact on energy and protein metabolism in forage-fed goats. Front. Vet. Sci. 2023, 10, 1272835. [Google Scholar] [CrossRef] [PubMed]
  29. Mirzaei, R.; Afaghi, A.; Babakhani, S.; Sohrabi, M.R.; Hosseini-Fard, S.R.; Babolhavaeji, K.; Ali Akbari, S.K.; Yousefimashouf, R.; Karampoor, S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed. Pharmacother. 2021, 139, 111619. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, J.; Xie, Y. Review on microbial degradation of zearalenone and aflatoxins. Grain Oil Sci. Technol. 2020, 3, 117–125. [Google Scholar] [CrossRef]
  31. Rais, A.; Chand, P.; Malik, M.; Prasad, T. Human microbiota: Role in cancer progression and therapy. In Microbial Crosstalk with Immune System; Academic Press: New Delhi, India, 2002; pp. 145–175. [Google Scholar]
  32. Hernández-Ramírez, J.O.; Nava-Ramírez, M.J.; Merino-Guzmán, R.; Téllez-Isaías, G.; Vázquez-Durán, A.; Méndez-Albores, A. The effect of moderate-dose aflatoxin B 1 and Salmonella Enteritidis infection on intestinal permeability in broiler chickens. Mycotoxin Res. 2020, 36, 31–39. [Google Scholar] [CrossRef] [PubMed]
  33. Adeleye, A.O.; Ajiboye, T.O.; Iliasu, G.A.; Abdussalam, F.A.; Balogun, A.; Ojewuyi, O.B.; Yakubu, M.T. Phenolic extract of Dialium guineense pulp enhances reactive oxygen species detoxification in aflatoxin B1 hepatocarcinogenesis. J. Med. Food 2014, 17, 875–885. [Google Scholar] [CrossRef] [PubMed]
  34. Cao, W.; Yu, P.; Yang, K.; Cao, D. Aflatoxin B1: Metabolism, toxicology, and its involvement in oxidative stress and cancer development. Toxicol. Mech. Methods 2022, 32, 395–419. [Google Scholar] [CrossRef] [PubMed]
  35. Olayem, B.S.; Olaitan, O.B.; Akinola, A.B. Immunomodulatory plant based foods, it’s chemical, biochemical and pharmacological approaches. In Medicinal Plants-Chemical, Biochemical, and Pharmacological Approaches; IntechOpen: London, UK, 2023. [Google Scholar] [CrossRef]
  36. Zhang, Y.; Usman, S.; Li, Q.; Li, F.; Zhang, X.; Nussio, L.G.; Guo, X. Effects of antioxidant-rich Lactiplantibacillus plantarum inoculated alfalfa silage on rumen fermentation, antioxidant and immunity status, and mammary gland gene expression in dairy goats. J. Anim. Sci. Biotechnol. 2024, 15, 9. [Google Scholar] [CrossRef] [PubMed]
  37. Li, Z.; Sang, R.; Feng, G.; Feng, Y.; Zhang, R.; Yan, X. Microbiological and metabolic pathways analysing the mechanisms of alfalfa polysaccharide and sulfated alfalfa polysaccharide in alleviating obesity. Int. J. Biol. Macromol. 2024, 263, 130334. [Google Scholar] [CrossRef] [PubMed]
  38. Amir, A.; McDonald, D.; Navas-Molina, J.A.; Kopylova, E.; Morton, J.T.; Zech Xu, Z.; Kightley, E.P.; Thompson, L.R.; Hyde, E.R.; Gonzalez, A.; et al. Deblur rapidly resolves single-nucleotide community sequence patterns. mSystems 2017, 2, e00191–e00216. [Google Scholar] [CrossRef] [PubMed]
  39. Baxter, M.F.; Merino-Guzman, R.; Latorre, J.D.; Mahaffey, B.D.; Yang, Y.; Teague, K.D.; Graham, L.E.; Wolfenden, A.D.; Hernandez-Velasco, X.; Bielke, L.R.; et al. Optimizing fluorescein isothiocyanate dextran measurement as a biomarker in a 24-h feed restriction model to induce gut permeability in broiler chickens. Front. Vet. Sci. 2017, 4, 56. [Google Scholar] [CrossRef] [PubMed]
  40. Paulson, J.N.; Stine, O.C.; Bravo, H.C.; Pop, M. Differential abundance analysis for microbial marker-gene surveys. Nat. Methods 2013, 10, 1200–1202. [Google Scholar] [CrossRef] [PubMed]
  41. McMurdie, P.J.; Holmes, S. phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 2013, 8, e61217. [Google Scholar] [CrossRef] [PubMed]
  42. Oksanen, J.; Blanchet, F.G.; Kindt, R.; Legendre, P.; Minchin, P.R.; O’hara, R.; Simpson, G.L.; Solymos, P.; Stevens, M.H.H.; Wagner, H. Vegan: Community Ecology Package, Package Version 2.5-5. 2019. Available online: https://github.com/vegandevs/vegan (accessed on 6 May 2024).
  43. Segata, N.; Izard, J.; Waldron, L.; Gevers, D.; Miropolsky, L.; Garrett, W.S.; Huttenhower, C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011, 12, R60. [Google Scholar] [CrossRef] [PubMed]
  44. SAS Institute Inc. SAS/Share: 9.4 User’s Guide, 2nd ed.; SAS Documentation: Cary, NC, USA, 2002; 288p. [Google Scholar]
Figure 1. Alpha and beta diversities of the cecal microbiota among different experimental groups. The cecal contents (n = 7/treatment) were subjected to 16S rRNA gene sequencing. Observed ASV, Pielou’s Evenness, and Shannon Index were calculated to measure the α-diversity of the cecal microbiota. Kruskal–Wallis test was used for statistical significance determination. The β-diversity-weighted UniFrac and unweighted UniFrac distances were used to generate the principal coordinates analysis (PCoA) plots. Permutational multivariate analysis of variance (PERMANOVA) was used for statistical significance determination. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Figure 1. Alpha and beta diversities of the cecal microbiota among different experimental groups. The cecal contents (n = 7/treatment) were subjected to 16S rRNA gene sequencing. Observed ASV, Pielou’s Evenness, and Shannon Index were calculated to measure the α-diversity of the cecal microbiota. Kruskal–Wallis test was used for statistical significance determination. The β-diversity-weighted UniFrac and unweighted UniFrac distances were used to generate the principal coordinates analysis (PCoA) plots. Permutational multivariate analysis of variance (PERMANOVA) was used for statistical significance determination. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Ijms 25 07977 g001
Figure 2. Differential enrichment of bacterial ASVs between different experimental groups (n = 7/treatment) was determined using linear discriminant analysis (LDA) effect size (LEfSe), with the all-against-all multiclass analysis, p < 0.05, and a logarithmic LDA threshold of 3.0. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Figure 2. Differential enrichment of bacterial ASVs between different experimental groups (n = 7/treatment) was determined using linear discriminant analysis (LDA) effect size (LEfSe), with the all-against-all multiclass analysis, p < 0.05, and a logarithmic LDA threshold of 3.0. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Ijms 25 07977 g002
Figure 3. Relative abundances of differentially enriched bacterial ASVs (n = 7/treatment). Significance was calculated using Kruskal–Wallis test. a,b indicates significant differences between treatments (p < 0.05). Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Figure 3. Relative abundances of differentially enriched bacterial ASVs (n = 7/treatment). Significance was calculated using Kruskal–Wallis test. a,b indicates significant differences between treatments (p < 0.05). Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Ijms 25 07977 g003
Figure 4. Morphometric analysis of the ileum in turkey poults fed a diet containing AFB1 and adsorbent materials. Histological images were taken using a 4× objective on H&E-stained tissue sections. (A) Control, AFB1-free diet; (B) AF, AFB1-contaminated diet at 250 ng/g; (C) alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; (D) alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and (E) YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group). L = Large (µm); W = Width (µm); A = Area (µm2).
Figure 4. Morphometric analysis of the ileum in turkey poults fed a diet containing AFB1 and adsorbent materials. Histological images were taken using a 4× objective on H&E-stained tissue sections. (A) Control, AFB1-free diet; (B) AF, AFB1-contaminated diet at 250 ng/g; (C) alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; (D) alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and (E) YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group). L = Large (µm); W = Width (µm); A = Area (µm2).
Ijms 25 07977 g004
Table 1. Relative abundances (%) of the cecal bacterial phyla and families among the experimental groups.
Table 1. Relative abundances (%) of the cecal bacterial phyla and families among the experimental groups.
TaxonControlAFAlfalfaAlfalfa + AFAF + YCWSEMp-Value
Phyla
Firmicutes79.4274.0481.0282.4572.781.920.13
Proteobacteria14.1020.7616.5212.9320.821.640.17
Tenericutes3.852.841.112.944.570.580.45
Cyanobacteria0.781.450.080.260.020.260.62
Actinobacteria0.450.080.080.420.150.080.39
Families
Oscillospiraceae32.7232.5340.0539.9030.042.060.06
Lachnospiraceae32.2825.6928.6628.6428.211.050.15
Enterobacteriaceae14.1020.7516.5212.8920.821.640.17
Clostridiales_unidentified4.855.974.113.294.050.450.24
Erysipelotrichaceae3.811.704.742.544.040.540.09
Bacillaceae1.551.470.524.202.120.610.06
Mollicutes_unidentified3.042.560.812.194.010.520.30
Lactobacillaceae1.671.840.361.511.740.270.10
Christensenellaceae0.760.810.800.931.230.080.68
Streptococcaceae0.09 b0.97 a1.09 a0.71 a0.45 a0.180.04
Vampirovibrio_unidentified0.781.450.080.260.020.260.62
Enterococcaceae0.670.140.100.200.230.100.18
Clostridia_unidentified0.140.850.03000.160.58
Peptostreptococcaceae0.410.290.090.130.090.060.12
Clostridiaceae 10.250.290.190.120.190.020.69
The mean relative abundances (%) of top five phyla and top 15 families of cecal microbiota of different groups are shown means (n = 7/treatment). a,b indicates significant differences between the treatments within the rows (p < 0.05). Statistical as significance was determined using the Kruskal–Wallis test, followed by the pairwise Wilcoxon rank sum test. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Table 2. Relative abundances (%) of the cecal bacterial genera and ASVs among the experimental groups.
Table 2. Relative abundances (%) of the cecal bacterial genera and ASVs among the experimental groups.
TaxonControlAFAlfalfaAlfalfa + AFAF + YCWSEMp-Value
Genera
Escherichia/Shigella13.1620.7116.3712.7620.671.730.17
Oscillospiraceae_unidentified10.2111.1410.9714.1111.050.670.68
Mediterraneibacter10.547.508.769.498.960.490.62
Lachnospiraceae_unidentified6.276.076.297.306.830.220.69
Subdoligranulum8.704.937.296.653.030.980.59
Pseudoflavonifractor5.045.914.367.055.760.45 0.32
Enterocloster4.643.545.245.053.990.320.48
Clostridiales_unidentified4.855.974.113.294.050.450.24
Blautia4.242.672.361.562.370.440.21
Bacillus1.551.470.524.202.120.610.06
Faecalibacterium0.180.267.700.370.661.460.07
Coprobacillaceae_unidentified1.72 ab0.44 b3.06 a0.69 b2.24 a0.480.02
Mollicutes_unidentified3.042.560.812.194.010.520.30
Anaerostipes2.510.751.351.431.330.280.11
Eisenbergiella1.501.551.230.781.440.140.46
ASVs
Escherichia/Shigella_F112.820.2415.8612.5420.231.690.14
Subdoligranulum_variabile_F45.111.883.803.361.930.600.81
Mediterraneibacter_F22.652.021.372.543.790.390.25
Mediterraneibacter_F32.143.313.411.022.03 0.440.35
Enterocloster_F52.651.252.562.082.060.240.44
Bacillus_F81.551.470.524.202.120.610.06
Mollicutes_unidentified_F92.771.190.640.803.300.540.50
Blautia_obeum_F182.931.671.341.071.310.330.82
Coprobacillaceae_unidentified_F131.72 ab0.44 b3.06 a0.69 b2.24 a0.480.02
Enterocloster_F111.241.361.511.681.040.100.79
Pseudoflavonifractor_capillosus_F160.931.221.031.302.430.270.45
Pseudoflavonifractor_F141.101.571.171.481.290.08 0.57
Oscillospiraceae_unidentified_F170.800.921.061.391.960.200.98
Oscillospiraceae_unidentified_F232.080.330.781.091.730.310.30
Pseudoflavonifractor_F201.351.140.552.060.440.290.21
Butyricicoccus_pullicaecorum_F241.290.131.721.980.470.350.68
Anaerostipes_butyraticus_F191.770.561.200.711.100.210.18
Faecalibacterium_F70.13 b0.24 b4.60 a0.34 b0.36 b0.860.02
Pseudoflavonifractor_F210.821.070.731.251.230.100.60
Oscillibacter_F150.921.250.581.300.910.130.73
The mean relative abundances (%) of top 15 genera, and top 20 ASVs of cecal microbiota of different groups are shown as means (n = 7/treatment). a,b indicates significant differences between the treatments within the rows (p < 0.05). Statistical significance was determined using the Kruskal–Wallis test, followed by the pairwise Wilcoxon rank sum test. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
Table 3. Effect of powdered alfalfa leaves on ileum morphometric analysis §, serum levels of FITC-d ¥, and cutaneous basophil hypersensitivity response (CBH) in turkey poults consuming a maize–soybean-meal-based diet contaminated with 250 ng AFB1/g for 28 days.
Table 3. Effect of powdered alfalfa leaves on ileum morphometric analysis §, serum levels of FITC-d ¥, and cutaneous basophil hypersensitivity response (CBH) in turkey poults consuming a maize–soybean-meal-based diet contaminated with 250 ng AFB1/g for 28 days.
ControlAFAlfalfaAlfalfa + AFAF + YCWSEM *p-Value
Villus height (μm)780.0 c350.6 a693.3 bc611.9 b700.0 bc271.8<0.0001
Villus width (μm)118.9 a146.3 b108.8 a112.7 a116.9 a36.90.02
Total area (μm2)103.2 c44.7 a71.7 b63.6 b74.1 b33.3<0.001
FITC-d (ng/mL)263.3 b858.2 a214.8 b213.5 b230.7 b654.70.007
CBH (mm)0.37 b0.50 b0.53 b0.86 a0.96 a0.23<0.0001
a,b,c Means with non-matching superscripts within rows indicates significant difference at p < 0.05. § Sixty-three measurements were taken per variable (In each treatment). ¥ Seven replicates/group (n = 1 poults per replicate). * Standard error of the mean. Control, AFB1-free diet; AF, AFB1-contaminated diet at 250 ng/g; alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; alfalfa + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) adsorbent; and YCW + AF, AFB1-contaminated diet at 250 ng/g + 0.5% (w/w) commercial yeast cell wall-based adsorbent (reference group).
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

Nava-Ramírez, M.d.J.; Liu, J.; Hernández-Ramírez, J.O.; Hernandez-Velasco, X.; Latorre, J.D.; Vázquez-Durán, A.; Zhang, G.; Senas-Cuesta, R.; Gómez-Rosales, S.; Stein, A.; et al. Exploring the Effects of an Alfalfa Leaf-Derived Adsorbent on Microbial Community, Ileal Morphology, Barrier Function, and Immunity in Turkey Poults during Chronic Aflatoxin B1 Exposure. Int. J. Mol. Sci. 2024, 25, 7977. https://doi.org/10.3390/ijms25147977

AMA Style

Nava-Ramírez MdJ, Liu J, Hernández-Ramírez JO, Hernandez-Velasco X, Latorre JD, Vázquez-Durán A, Zhang G, Senas-Cuesta R, Gómez-Rosales S, Stein A, et al. Exploring the Effects of an Alfalfa Leaf-Derived Adsorbent on Microbial Community, Ileal Morphology, Barrier Function, and Immunity in Turkey Poults during Chronic Aflatoxin B1 Exposure. International Journal of Molecular Sciences. 2024; 25(14):7977. https://doi.org/10.3390/ijms25147977

Chicago/Turabian Style

Nava-Ramírez, María de Jesús, Jing Liu, Juan Omar Hernández-Ramírez, Xochitl Hernandez-Velasco, Juan D. Latorre, Alma Vázquez-Durán, Guolong Zhang, Roberto Senas-Cuesta, Sergio Gómez-Rosales, Andressa Stein, and et al. 2024. "Exploring the Effects of an Alfalfa Leaf-Derived Adsorbent on Microbial Community, Ileal Morphology, Barrier Function, and Immunity in Turkey Poults during Chronic Aflatoxin B1 Exposure" International Journal of Molecular Sciences 25, no. 14: 7977. https://doi.org/10.3390/ijms25147977

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