Skip to main content

Persea americana extract protects intestinal tissue from Eimeria papillata-induced murine Infection

Abstract

Coccidiosis is the most prevalent disease-causing widespread economic loss among farm and domestic animals. Currently, several drugs are available for the control of this disease but resistance has been confirmed for all of them. There is an urgent need, therefore, for the identification of new sources as alternative treatments to control coccidiosis. The present work aimed to study the effect of the Persea americana extract (PAE) as an anti-coccidial, anti-oxidant, and anti-apoptotic modulator during murine intestinal Eimeria papillata infection. A total of 25 male mice were divided into five groups, as follows: Group1: Non-infected-non-treated (negative control), Group2: Non-infected-treated group with PAE (500 mg/kg b.w). Group3: Infected-non-treated (positive control), Group4: Infected-treated group with PAE (500 mg/kg b.w.), and Group5: Infected-treated group with Amprolium (120 mg/kg b.w.). Groups (3–5) were orally inoculated with 1 × 103 sporulated E. papillata oocysts. After 60 min of infection, groups (4 and 5) were treated for 5 consecutive days with the recommended doses of PAE and amprolium. The fact that PAE has an anti-coccidial efficacy against intestinal E. papillata infection in mice has been clarified by the reduction of fecal oocyst output on the 5th day post-infection by about 85.41%. Moreover, there is a significant reduction in the size of each parasite stage in the jejunal tissues of the infected-treated group with PAE. PAE counteracted the E. papillata-induced loss of glutathione peroxidase (GPx), superoxide dismutase (SOD), and total antioxidant capacity (TCA). E. papillata infection also induced an increase in the apoptotic cells expressed by caspase-3 which modulated after PAE treatment. Moreover, the mRNA expression of the goblet cell response gene, mucin (MUC2), was upregulated from 0.50 to 1.20-fold after treatment with PAE. Based on our results, PAE is a promising medicinal plant with anti-coccidial, anti-oxidant, and anti-apoptotic activities and could be used as a food additive.

Peer Review reports

Introduction

Intestinal coccidiosis is a cosmopolitan disease affecting a wide variety of vertebrates [1, 2]. The causative agent for this protozoan disease is the apicomplexan species within the genus Eimeria (family Eimeriidae). Ernst et al. [3] identified Eimeria papillata as a coccidian parasite in the house mouse Mus musculus. Infection with this protozoan parasite occurs via the fecal-oral route of oocysts with a high degree of host specificity [4]. The most commonly used method for the detection of coccidia is the flotation technique of oocysts shed in feces [5]. This parasite species, E. papillata, spends its life cycle within the intestinal tract causing extensive damage to the intestinal mucosa, inflammation, and oxidative stress that affects general body performance [6, 7].

Therapeutic tools for coccidiosis have relied on the availability of more than 30 anticoccidial drugs [8, 9]. However, the intensive use of these drugs has led to side effects on animal health and the development of drug-resistant Eimeria strains [10]. Researchers’ efforts are now directed toward finding alternative agents with no side effects on the host infected with Eimeria species [11]. Among, other available options, different compounds obtained from botanicals have shown excellent and admirable anticoccidial and other therapeutic effects [12]. In Saudi Arabia, natural sources such as Allium sativum [13, 14], Phoenix dactylifera [15], Punica granatum [16], Ziziphus spina-christi [17], Salvadora persica [18,19,20], Morus nigra [21], Zingiber officinale [22, 23], and Azadirachta indica [24,25,26] have been evaluated as alternative controls to murine coccidiosis.

Persea americana, also known as aguacate (avocado), belongs to the family Lauraceae. The fruits are edible, while the bark, leaves, stem, and roots are utilized as a local remedy [27]. Phytochemical analysis of avocados has revealed a variety of bioactive compounds including phenolics, flavonoids, carotenoids, tannins, saponins, alkaloids, vitamin C, and vitamin E [28, 29]. The medicinal properties attributed to P. americana include anti-hypertensive [30], hepatoprotective [31], anti-ulcer [32], anti-cancer [33], insecticidal [34, 35], anti-microbial [36,37,38,39,40], anti-oxidant [41], antidiabetic [42, 43], anti-inflammatory [44,45,46], and anti-coccidial properties [47].

In this study, the role of P. americana extract was investigated against the expression of the cysteine aspartic acid protease-3 (caspase-3), the goblet cells regulating gene, and the oxidative damage caused by E. papillata infection in mouse jejunum.

Materials and methods

Preparation of the avocado peel extract

Persea americana (avocado) fruits were purchased from the local markets in Riyadh, Saudi Arabia. Edible pulps were removed, cut into pieces, air-dried at 40ºC, and then pulverized using an electrical grinder. The obtained powder (100 g) was macerated using 1000 ml methanol (70%) for 42 h with vigorous shaking. The methanolic P. americana extract (PAE) was filtered and evaporated under reduced pressure [16]. PAE was dissolved in distilled H2O to be used for experimental steps.

Determination of phenolic and flavonoid contents

The total phenolic content was determined using the Folin–Ciocalteu technique as described by Abdel Moneim [48]. Absorbance was measured at 760 nm with a spectrophotometer (PD 303 UV spectrophotometer, Apel Co., Limited, Saitama, Japan). The measured value was compared to a calibration curve built with gallic acid solutions, and the results are given as mg gallic acid per gram of dry extract (mg GAE/g). Moreover, the total flavonoid content was determined using the aluminum chloride colorimetric method of Abdel Moneim [48]. Absorbance at 510 nm was measured. The flavonoid value was calculated using a calibration curve and reported as mg quercetin per gram dry extract (mg QE/g).

The 2,2-Diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity

The activity of PAE was determined to scavenge DPPH radicals according to Akillioglu and Karakaya [49]. Absorbance was measured at 515 nm using a microplate reader (ELX 800; Bio-Tek Instruments, Winooski, VT, USA). The antioxidant activity is expressed as suppression % of DPPH radicals.

Passaging of Eimeria species

Eimeria papillata was used as a model murine coccidian parasite and obtained from Prof Heinz Mehlhorn (Heinrich-Heine-Universitat, Germany). Five laboratory mice (Mus musculus) were obtained from the animal house at the Department of Zoology (College of Science, King Saud University) and inoculated with 1 × 103 sporulated E. papillata oocysts by oral gavage. On the 5th day post-infection (p.i.), feces were collected and sporulated in 2.5% (w/v) potassium dichromate (K2Cr2O7) at room temperature [50]. The sporulated oocysts were washed in a phosphate buffer solution (PBS) and used in this experiment. Using an Olympus B×61 microscope (Tokyo, Japan), oocysts (sporulated and non-sporulated) were photographed and described using the guidelines of Duszynski and Wilber [51].

Experimental design

Twenty-five male C57BL/6 mice (10–12 weeks) were obtained from the College of Pharmacy at King Saud University. All mice have been bred under specified pathogen-free conditions and fed a standard diet and water ad libitum. Mice were divided into five groups (5 mice/group), as follows: Group1: Non-infected-non-treated (negative control), Group2: Non-infected-treated group with PAE (500 mg/kg b.w). Group3: Infected-non-treated (positive control), Group4: Infected-treated group with PAE (500 mg/kg b.w.), and Group5: Infected-treated group with Amprolium (120 mg/kg b.w.). Groups (3–5) were orally inoculated with 1 × 103 sporulated E. papillata oocysts in 100 µl of physiological saline. After 60 min of infection, groups (4 and 5) were treated for 5 consecutive days with the recommended doses of PAE and amprolium via oral gavage in 100 µl based on the previous study of Al-Otaibi et al. [47].

Sample collection

On the 5th day p.i., fresh fecal pellets of each mouse from all experimental groups were collected separately and examined for the presence of E. papillata oocysts. According to Schito et al. [52], the number of oocysts per gram of feces was estimated using the McMaster technique. Additionally, the suppression (%) of oocyst shedding was calculated as follows: 100 – (oocysts output in the treated group/oocysts output in the infected group) × 100.

Histological examination of parasite stages

On the 5th day p.i., CO2 asphyxia was used for the euthanasia of all experimental animals. To evaluate the morphometric changes among the Eimeria stages in the mouse jejunum, pieces of jejuna were collected after dissection on the 5th -day p.i. of mice and fixed in formalin (10%) for 24 h, dehydrated and embedded in paraffin wax. Sections were cut and stained with hematoxylin and eosin (H&E) [53]. Under an Olympus B×61 microscope (Tokyo, Japan), parasite stages (gamonts and developing oocysts) were observed in sections of the infected and infected-treated groups and then measured using a calibrated ocular micrometer.

Oxidative status in the jejunum

Parts of jejunumwere weighed and homogenized in an ice-cold medium of 50 mM Tris-HCl and 300 mM sucrose. The mixture was centrifuged for 10 min (500×g and 4 ºC) to give a final yield of 10% (w/v) jejunal homogenate and then kept at -20 °C until use [54]. For different biochemical assays, the supernatant was used and evaluated colorimetrically to determine glutathione peroxidase (GPx) [55], superoxide dismutase (SOD) [56], and total antioxidant capacity (TAC) [57] with its related kits (Biodiagnostic Co., Egypt). The absorbance of the reactions was measured by Molecular Device (Spectra MAX 190) provided with SoftMax® Pro software v. 6.3.1.

Immunohistochemical staining of Caspase-3

Paraffin-embedded jejunal sections were treated with 3% H2O2 for 10 min, blocked with fetal bovine serum (5%), and then incubated at 4 °C overnight with a primary polyclonal rabbit anti-mouse antibody specific for cysteine aspartic acid protease-3 (Caspase-3) (1:100 dilution in PBS, Santa Cruz Biotechnology, CA, USA), according to Dkhil et al. [58]. After triplicate washing with PBS, samples were treated with a biotin-conjugated secondary antibody (1:2,000 dilution in PBS). Sections were counterstained for 1 min with hematoxylin and re-incubated for 15 min with streptavidin which was labeled with horseradish peroxidase. All sections were photographed using an Olympus B×61 microscope (Tokyo, Japan).

Goblet cell response gene (MUC2) expression

Using Trizol (Invitrogen), total RNA was isolated from the preserved samples (at -80 °C). RNA samples were treated with DNase (Applied Biosystems, Darmstadt, Germany) for at least 1 h and then converted into cDNA using the reverse transcription kit (Qiagen, Hilden, Germany) following the manufacturer’s procedure. Quantitative real-time PCR (qRT-PCR) was performed using the ABI Prism® 7500HT sequence detection system (Applied Biosystems, Darmstadt, Germany) with QuantiTect™ SYBR® green PCR master mix (Qiagen, Hilden, Germany) and the gene-specific primers (Qiagen, Hilden, Germany): goblet cell response gene (MUC2) (Mm_Muc2_2_SG, Cat. No. Mm_Muc2_2_SG) and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (Mm_Gapdh_3_SG, Cat. No. QT01658692). The 2−ΔΔCT method of Livak and Schmittgen [59] was used to evaluate the fold-change in mRNA expression. GAPDH was used as a reference gene.

Statistical analysis

Differences between obtained values (mean ± SD) for experimental groups were compared by two-way analysis of variance (ANOVA) using SigmaPlot® version 11.0 (Systat Software, Inc., Chicago, IL, USA). The p-value ≤ 0.05 was considered a statistically significant difference.

Results

The total phenolic content in PAE was determined using the Folin–Ciocalteu technique as 111.8 ± 1.38 mg GAE/g (Fig. 1). Moreover, the total flavonoid content in PAE was determined using the aluminum chloride colorimetric method as 67.63 ± 4.85 mg QE/g (Fig. 1). The DPPH radical scavenging activity was 85.53 ± 1.58 for PAE.

Fig. 1
figure 1

The concentration of phenolics (mg GAE/g) and flavonoids (mg QE/g) in PAE

Mice in the experimental groups (3–5) started shedding oocysts (non-sporulated) after 3 days p.i. On the 5th day p.i., the Eimeria oocyst output was observed to be 4.075 × 109 oocysts/g feces in the infected group, which is associated with general weakness, poor body performance, loss of appetite, and diarrhea. PAE was significantly able to suppress the oocyst output by 85.41% in comparison to 80.38% in the drug-treated group (Fig. 2). Oocysts were sub-spherical and surrounded by a thick bi-layered wall (Fig. 3). After sporulation, four ellipsoidal sporocysts were observed with two sporozoites per each (Fig. 3).

Fig. 2
figure 2

Suppression rate of E. papillata oocysts in the infected and infected-treated mice with PAE and AMP groups. Significance at p ≤ 0.05 against the infected group (*)

Fig. 3
figure 3

Eimeria papillata oocyst A non-sporulated oocyst. B sporulated oocyst. C oocyst bi-layered. D site of splitting sporocysts during excystation (black arrow). Scale bar = 10 μm (A, B, D), and 5 μm (C), (OM Outer membrane, IM Inner membrane of oocyst, Z Zygote, SP Sporocysts, SPZ Sporozoites)

The developmental Eimeria stages appeared inside the jejunal tissue (Fig. 4). The reduction of oocyst output was due to the impaired development of Eimeria stages. In the infected group, microgamonts measured 17.94 ± 0.18 μm, macrogamonts 17.26 ± 0.41 μm, and developing oocysts 18.18 ± 0.13 μm (Table 1). After treatment with PAE, there was a significant morphometrical reduction to 16.67 ± 0.08 μm (microgamonts), 12.87 ± 0.29 μm (macrogamonts), and 15.00 ± 0.19 μm (developing oocysts) in comparison to the drug-treated group.

Fig. 4
figure 4

Histology of jejunal tissue of mice in different experimental groups. A E. papillata infected jejunum with an increased number of parasite stages. B and C infected treated mouse (PAE and AMP, respectively) with decreased number of parasite stages. MI Microgamonts, MA Macrogamonts, ME Merozoites, DO Developing oocyst). Scale bar = 50 μm

Table 1 Morphometric changes of developmental changes of Eimeria papillata in infected and treated groups

The GPx level significantly declined from 24.31 ± 5.22 in the non-infected group to 15.80 ± 1.79 mg/g tissue in the infected group. While, the level of GPx of mice treated with PAE and reference drug was elevated to 24.80 ± 1.40 and 20.42 ± 3.44 mg/g tissue, respectively (Fig. 5). Moreover, the SOD level significantly declined from 6.38 ± 0.25 in the non-infected group to 3.18 ± 0.46 U/g tissue in the infected group. While, the level of SOD in mice treated with PAE and reference drug was significantly elevated to 4.72 ± 0.48 and 4.95 ± 0.41 U/g tissue, respectively (Fig. 5). There was a significant decline in the level of TAC from 0.42 ± 0.01 in the non-infected group to 0.28 ± 0.04 mM/L in the infected group. While, TAC of mice treated with PAE and reference drug was significantly elevated to 0.37 ± 0.02 and 0.37 ± 0.01 mM/L, respectively (Fig. 5).

Fig. 5
figure 5

Effect of PAE on glutathione peroxidase, superoxide dismutase, and total antioxidant levels in mouse jejunum infected with E. papillata.*Significance changes concerning the control group,#Significance changes for the infected group

The role of PAE in Eimeria infection-induced apoptosis was checked, through the histochemical staining for caspase-3 in the mice jejuna from different experimental groups. Infection with E. papillata induced apoptotic changes within the jejunal tissues of the infected mice group (Fig. 6). Immunohistochemical investigation for caspase-3 showed that PAE was able to decrease the immunoreactivity in the jejuna of mice infected with E. papillata (Fig. 6).

Fig. 6
figure 6

Immunohistochemical localization of caspase-3 in the jejuna of mice. A control non-infected jejunum. B E. papillata infected jejunum with an increased number of caspase-3 positive cells. C and D infected treated mouse (PAE and AMP, respectively) with decreased number of caspase-3 positive cells. Scale bar = 50 μm

qRT-PCR revealed downregulation in the expression level of the MUC2 gene in the mice jejunum (at 5th -day p.i.) due to E. papillata infection (Fig. 7). However, treatment with PAE significantly upregulated the MUC2 gene expression from 0.50 to 1.20-fold (Fig. 7). Data were normalized to the GAPDH mRNA level and shown as fold induction (in log 2 scale) relative to the mRNA level in the control by RT-PCR.

Fig. 7
figure 7

Effect PAE on the mRNA expression of MUC2 in the jejunal samples from E. papillata-infected mice. The expression values obtained by RT-PCR analysis were normalized to the reference gene GAPDH mRNA level and are shown as fold induction (in log 2 scale) relative to the mRNA level in the control. *Significance changes concerning the control group, #Significance changes concerning the infected group

Discussion

Since ancient times, natural sources including plants have been used in the treatment of various diseases. Avocado is one of the natural sources which have a chemoprotective effect [60,61,62]. This study showed that PAE is an efficient agent in ameliorating E. papillata infection in mice as it could reduce the oocyst shedding rate on 5th -day p.i. by about 85.41%. This inhibition of Eimeria infection is known to occur with most anticoccidial drugs. This diminished output suggests that PAE impairs the development of intracellular Eimeria stages in the host intestinal cells before the relatively inert oocyst is formed and finally released. The fact that PAE possesses anti-coccidial activity has been previously reported by Al-Otaibi et al. [47] regard to the content of the extract. Ferreira da Vinha et al. [63], and Rahman et al. [29] found that the PAE contains phenolics, flavonoids, carotenoids, tannins, saponins, alkaloids, and vitamins. Significant changes in the size of Eimeria stages were observed after PAE treatment. This might be due to the polyphenolic compounds of PAE which exert antimicrobial activity leading to impaired membrane functions and leakage of cellular constituents [64].

Our findings demonstrated E. papillata infection is associated with oxidative damage to the mice jejunum, which leads to the depletion of antioxidant enzymes and reduction of GPx, SOD, and TAC which are indispensable for protecting the animal body from the damage caused by free radicals during Eimeria infection. Previous studies [14, 15, 63, 65,66,67] reported that the imbalance of the antioxidant defense system due to Eimeria infection leads to harmful cellular effects. The treatment of E. papillata-infected mice with PAE significantly resulted in the pronounced modulation of oxidative damage and enhanced antioxidant capacity in the jejunum of mice. These results showed that PAE acts as an excellent antioxidant activity, agreed with Vo et al. [68] stated the presence of free radical scavenging properties in avocados which offer protection against oxidative damage. In our previous study, we proved that glutathione reduced (GSH), nitric oxide (NO), and malondialdehyde (MDA) activities improved in the jejuna of E. papillata-infected mice after treatment with PAE due to the presence of phenolic compounds [47].

Previous studies by Lüder et al. [69] and Balamurugan et al. [70] reported that apoptosis could regulate the host response to a variety of intracellular parasitic infections and help to eliminate the infected cells. Alkhudhayri et al. [71] studied the relationship between the developmental stages of E. papillata and host apoptosis. In this study, the death of jejunal cells in the infected mice was evidenced by a significant observation of the pro-apoptotic markers of caspase-3. This agreed with Dkhil et al. [24], Metwaly et al. [15], and Abdel-Gaber et al. [25], who reported that parasite invasion and replication may cause considerable stress to the host cells which triggered apoptosis for the infected intestinal cells. Treatment of infected mice with PAE significantly reduced the rate of caspase-3 and improved the apoptotic changes in jejunal cells, this agreed with those stated the anti-apoptotic activity of avocado extracts of Bonilla-Porras et al. [72], Abozaid et al. [73], and El-Magd et al. [74].

Goblet cells (GCs) are considered a dynamic protective agent against pathogens [75]. GCs are produced from stem cells (SCs) that are confined to the intestinal crypts [76]. Inside GCs, the MUC2 gene is widely expressed and is responsible for the regulation of mucin secretion and inflammatory response in preventing pathogen-induced epithelial injury [18, 24, 77]. Our results of qRT-PCR revealed that the expression of the MUC2 gene was significantly downregulated in the mice jejuna causing physical contact between E. papillata and host cells, which is consistent with previous studies [16, 18, 24, 25, 66, 78, 79]. This result reflects that, during infection, SCs are parasitized and become unable to produce GCs associated with the downregulation of the MUC2 gene. Previous studies [80,81,82] reported that the alteration in goblet cells could affect the susceptibility of the Eimeria-infected host to limit the capacity of the parasite to penetrate the epithelial cells. PAE, based on our results, was able to alter this downregulation of MUC2 due to infection. The fact that PAE is effective in ameliorating the upregulation of genes associated with inflammation. This agreed with Al-Otaibi et al. [47] mentioned that PAE has a role in the regulation of goblet cell-producing mucin which helps to improve the inflammatory response to infectious diseases.

Conclusion

Our data indicate that avocados possess an anti-oxidant and anti-apoptotic activity against murine coccidiosis. PAE could be used with normal animal food as an additive to protect host tissue from injuries induced by various pathogenic infections.

Availability of data and materials

All the datasets generated or analyzed during this study are included in this published article.

References

  1. Dalloul RA, Lillehoj HS. Poultry coccidiosis: recent advancements in control measures and vaccine development. Expert Rev Vaccines. 2006;5(1):143–63.

    Article  CAS  PubMed  Google Scholar 

  2. Mehlhorn H. (2008) Encyclopedia of parasitology: Volume 1, AM. Encyclopedia of parasitology: Volume 1, AM., (Edn. 3).

  3. Ernst JV, Chobotar B, Hammond DM. The oocysts of Eimeria Vermiformis sp. n. and E. papillata sp. n. (Protozoa: Eimeriidae) from the mouse Mus musculus. J Protozool. 1971;18:221–3.

    Article  CAS  PubMed  Google Scholar 

  4. Wilber PG, Duszynski D, Upton S, Seville RS, Corliss JO. A revision of the taxonomy and nomenclature of the Eimeria spp. (Apicomplexa: Eimeriidae) from rodents in the Tribe Marmotini (Sciuridae). Syst Parasitol. 1998;39:113–35.

    Article  Google Scholar 

  5. Joachim A, Altreuther G, Bangoura B, Charles S, Daugschies A, Hinney B, Lindsay DS, Mundt H, Ocak M, Sotiraki S. WAAVP guideline for evaluating the efficacy of anticoccidials in mammals (pigs, dogs, cattle, sheep). Vet Parasitol. 2018;253:102–19.

    Article  PubMed  Google Scholar 

  6. Heitlinger E, Spork S, Lucius R, Dieterich C. The genome of Eimeria falciformis-reduction and specialization in a single host apicomplexan parasite. BMC Genom. 2014;15(1):1–17.

    Article  CAS  Google Scholar 

  7. Abdel-Latif M, Abdel-Haleem HM, Abdel-Baki AAS. Anticoccidial activities of Chitosan on Eimeria papillata-infected mice. Parasitol Res. 2016;115:2845–52.

    Article  PubMed  Google Scholar 

  8. Kant V, Singh P, Verma PK, Bais I, Parmar MS, Gopal A, Gupta V. Anticoccidial drugs used in the poultry: an overview. Sci Inter. 2013;1(7):261–5.

    Article  CAS  Google Scholar 

  9. Wunderlich F, Al-Quraishy S, Steinbrenner H, Sies H, Dkhil MA. Towards identifying novel anti-Eimeria agents: Trace elements, vitamins, and plant-based natural products. Parasitol Res. 2014;113:3547–56.

    Article  PubMed  Google Scholar 

  10. Chapman HD. Milestones in avian coccidiosis research: a review. Poult Sci. 2014;93(3):501–11.

    Article  CAS  PubMed  Google Scholar 

  11. Habibi H, Firouzi S, Nili H, Razavi M, Asadi SL, Daneshi S. Anticoccidial effects of herbal extracts on Eimeria tenella Infection in broiler chickens: in vitro and in vivo study. J Parasite Dis. 2016;40(2):401–7.

    Article  Google Scholar 

  12. Abbas A, Iqbal Z, Abbas RZ, Khan MK, Khan JA. In-vitro anticoccidial potential of Saccharum officinarum extract against Eimeria oocysts. Bol Latinoam Caribe Plant Med Aromat. 2015;14(6):456–61.

    Google Scholar 

  13. Al-Quraishy S, Delic D, Sies H, Wunderlich F, Abdel-Baki AZ, Dkhil MA. Differential miRNA expression in the mouse jejunum during garlic treatment of Eimeria papillata Infections. Parasitol Res. 2011;109(2):387–94.

    Article  PubMed  Google Scholar 

  14. Dkhil MA, Abdel-Baki AS, Wunderlich F, Al-Quraishy S. Anticoccidial and anti-inflammatory activity of garlic in murine Eimeria papillata Infections. Vet Parasitol. 2011;175(1):66–72.

    Article  CAS  PubMed  Google Scholar 

  15. Metwaly MS, Dkhil MA, Al-Quraishy S. Anti-coccidial and anti-apoptotic activities of palm pollen grains on Eimeria papillata-induced Infection in mice. Biologia. 2014;69(2):254–9.

    Article  Google Scholar 

  16. Amer OSO, Dkhil MA, Hikal WM, Al-Quraishy S. Antioxidant and anti-inflammatory activities of pomegranate (Punica granatum) on Eimeria papillata-induced infection in mice. BioMed Res Intern. 2015. https://0-doi-org.brum.beds.ac.uk/10.1155/2015/219670. (BioMed Res Inter 2015 (Article ID 219670), 7 pages).

    Article  Google Scholar 

  17. Alzahrani F, Al-Shaebi EM, Dkhil MA, AL-Quraishy S. In vivo anti-Eimeria and in vitro anthelmintic activity of Ziziphus spina-christi leaf extracts. Pakistan J Zool. 2016;48(2):409–13.

    CAS  Google Scholar 

  18. Thagfan FA, Dkhil MA, Al-Quraishy S. In vivo anticoccidial activity of Salvadora Persica root extracts. Pakistan J Zool. 2017;49(1):53–7.

    Google Scholar 

  19. Al-Quraishy S, Thagfan FA, Al-Shaebi EM, Qasem M, Abdel-Gaber R, Dkhil MAM. Salvadora Persica protects mouse intestine from eimeriosis. Braz J Vet Parasitol Jaboticabal. 2019;28(4):605–12.

    CAS  Google Scholar 

  20. Dkhil MA, Khalil MF, Diab MSM, Bauomy AA, Santourlidis S, Al-Shaebi EM, Al-Quraishy S. Evaluation of nanoselenium and nanogold activities against murine intestinal schistosomiasis. Saudi J Biol Sci. 2019;26:1468–72.

    Article  CAS  PubMed  Google Scholar 

  21. Thagfan FA, Al-Megrin WA, Al-Shaebi EM, Al-Quraishy S, Dkhil MA. Protective role of Morus nigra leaf extracts against murine Infection with Eimeria papillata. Comb Chem High Throughput Screen. 2021;24(10):1603–8.

    Article  CAS  PubMed  Google Scholar 

  22. Mubaraki MA, Thagfan FA, Alkhudhayri A, Al-Shaebi EM, Maodaa SN, Abdel-Gaber R, Hafiz TA, Al-Quraishy S, Dkhil MA. Zingiber officinale supplementation suppresses eimeriosis and regulates goblet cell response. Saudi J Biol Sci. 2022;29:3403–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dkhil MA, Thagfan FA, Morad MY, Al-Shaebi EM, Elshanat S, Bauomy AA, Mubaraki M, Hafiz TA, Al-Quraishy S, Abdel-Gaber R. Biosynthesized silver nanoparticles have anticoccidial and jejunum-protective effects in mice infected with Eimeria papillata. Environ Sci Pollut Res. 2023;30:44566–77.

    Article  CAS  Google Scholar 

  24. Dkhil MA, Al-Quraishy S, Abdel Moneim AE, Delic D. Protective effect of Azadirachta indica extract against Eimeria papillata-induced coccidiosis. Parasitol Res. 2012;112(1):101–6.

    Article  PubMed  Google Scholar 

  25. Abdel-Gaber R, Abu Hawsah M, Al-Otaibi T, Alojayri G, Al-Shaebi EM, Mohammed OB, Elkhadragy MF, Al-Quraishy S, Dkhil MA. Biosynthesized selenium nanoparticles to rescue coccidiosis-mediated oxidative stress, apoptosis and inflammation in the jejunum of mice. Front Immunol. 2023;14: 1139899.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Abdel-Gaber R, Abu Hawsah M, Al-Shaebi M, Al-Otaibi T, Thagfan FA, Al-Quraishy S, Dkhil MA. Effect of biosynthesized nanoselenium using Azadirachta indica (Meliaceae) leaf extracts against Eimeria papillata Infection. Microsc Res Tech. 2023b;86:714–24.

  27. -Ejiofor NC, Ezeagu IE, Ayoola M, Umera EA. Determination of the chemical composition of avocado (Persea americana) seed. AFTNSOJ. 2018;2:51–5.

    Google Scholar 

  28. Widiyastuti Y, Pratiwi R, Riyanto S, Wahyuono S. Cytotoxic activity and apoptosis induction of avocado Persea americana Mill. Seed extract on MCF-7 cancer cell line. Indones J Biotechnol. 2018;23(2):61–7.

    Article  Google Scholar 

  29. Rahman N, Sabang SM, Abdullah R, Bohari B. Antioxidant properties of the methanolic extract of avocado fruit peel (Persea americana Mill.) From Indonesia. J Adv Pharm Technol Res. 2022;13(3):166.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Adeboye JO, Fajonyomi MO, Makinde JM, Taiwo OB. A preliminary study on the hypotensive activity of Persea americana leaf extracts in anaesthetized normotensive rats. Fitoterapia. 1999;70(1):15–20.

    Article  Google Scholar 

  31. Kawagishi H, Fukumoto Y, Hatakeyama M, He P, Arimoto H, Matsuzawa T, Arimoto Y, Suganuma H, Inakuma T, Sugiyama K. Liver injury suppressing compounds from avocado (Persea americana). J Agric Food Chem. 2001;49(5):2215–21.

    Article  CAS  PubMed  Google Scholar 

  32. Ukwe CV, Nwafor SV. Anti-ulcer activity of aqueous leaf extract of Persea americana (family-Lauraceae). Nigerian J Pharm Res. 2004;3:91–5.

    Google Scholar 

  33. D’Ambrosio SM, Han C, Pan L, Kinghorn AD, Ding H. Aliphatic acetogenin constituents of avocado fruits inhibit human Oral cancer cell proliferation by targeting the EGFR/RAS/RAF/MEK/ERK1/2 pathway. Biochem Biophys Res Commun. 2011;409(3):465–9.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Leite JJ, Brito EH, Cordeiro RA, Brilhante RS, Sidrim JJ, Bertini LM, Morais SM, Rocha MF. Chemical composition, toxicity and larvicidal and antifungal activities of Persea americana (avocado) seed extracts. Rev Soc Bras Med Trop. 2009;42(2):110–3.

    Article  PubMed  Google Scholar 

  35. Torres RC, Garbo AG, Walde RZML. Larvicidal activity of Persea americana Mill. Against Aedes aegypti. Asian Pac J Trop Med. 2014;7:167-S170.

    Article  Google Scholar 

  36. De Almeida AP, Miranda MMFS, Simoni IC, Wigg MD, Lagrota MHC, Costa SS. Flavonol monoglycosides isolated from the antiviral fractions of Persea americana (Lauraceae) leaf infusion. Phytother Res. 1998;12(8):562–7.

    Article  CAS  Google Scholar 

  37. Rodríguez-Carpena JG, Morcuende D, Andrade MJ, Kylli P, Estévez M. Avocado (Persea americana Mill.) Phenolics, in vitro antioxidant and antimicrobial activities, and inhibition of lipid and protein oxidation in porcine patties. J Agric Food Chem. 2011;59(10):5625–35.

    Article  PubMed  Google Scholar 

  38. Lu YC, Chang HS, Peng CF, Lin CH, Chen IS. Secondary metabolites from the unripe pulp of Persea americana and their antimycobacterial activities. Food Chem. 2012;135(4):2904–9.

    Article  CAS  PubMed  Google Scholar 

  39. Guzmán VB, Silva DAO, Kawazoe U, Mineo JR. A comparison between IgG antibodies against Eimeria acervulina, E. maxima, and E. tenella and oocyst shedding in broiler-breeders vaccinated with live anticoccidial vaccines. Vaccine. 2003;21(27 30):4225–33.

    Article  PubMed  Google Scholar 

  40. Cardoso PF, Scarpassa JA, Pretto-Giordano LG, Otaguiri ES, Yamada-Ogatta SF, Nakazato G, Perugini MRE, Moreira IC, Vilas-Bôas GT. Antibacterial activity of avocado extracts (Persea americana Mill.) Against Streptococcus agalactiae. Inter J Exp Bot. 2016;85:218–24.

    Google Scholar 

  41. Adaramola B, Onigbinde A, Shokunbi O. Physiochemical properties and antioxidant potential of Persea americana seed oil. Chem Inter. 2016;2(3):168–75.

    CAS  Google Scholar 

  42. Anita BS, Okokon JE, Okon PA. Hypoglycemic activity of aqueous leaf extract of Persea americana Mill. Indian J Pharmacol. 2005;37:525–6.

    Google Scholar 

  43. Umoh IU, Aquaisua AN, Udo NM. The effect of fresh stem juice extract of Costus afer on the cytohistomorphology of the kidney in aspirin-treated Wistar rats. J Appl Biol Biotechnol. 2019;7(2):78–81.

    Article  CAS  Google Scholar 

  44. Ojewole JA, Amabeoku GJ. Anticonvulsant effect of Persea americana Mill (Lauraceae) (Avocado) leaf aqueous extract in mice. Phytother Res. 2006;20(8):696–700.

    Article  PubMed  Google Scholar 

  45. Adeyemi OO, Okpo SO, Ogunti OO. Analgesic and anti-inflammatory effects of the aqueous extract of leaves of Persea americana Mill (Lauraceae). Fitoterapia. 2002;73(5):375–80.

    Article  CAS  PubMed  Google Scholar 

  46. Hürkul MM, Sarıaltın SY, Köroğlu A, Çoban T. In vitro inhibitory potential of avocado fruits, Persea americana (Lauraceae) against oxidation, inflammation and key enzymes linked to Skin Diseases. Rev Biol Trop. 2021;69(2):472–81.

    Article  Google Scholar 

  47. Al-Otaibi T, Abu Hawsah M, Alojayri G, Mares MM, Aljawdah HMA, Maodaa SN, Al-Shaebi EM, Dkhil MA, Thagfan FA, Al-Quraishy S, Abdel-Gaber R. Vivo anticoccidial, antioxidant, and anti-inflammatory activities of avocado fruit, Persea americana (Lauraceae), against Eimeria papillata Infection. Parasitol Inter. 2023;95:102741.

    Article  CAS  Google Scholar 

  48. Abdel Moneim A. The neuroprotective effects of purslane (Portulaca oleracea) on rotenone-induced biochemical changes and apoptosis in brain of rat. CNS Neurol Disord Drug Targets. 2013;12(6):830–41.

    Article  CAS  PubMed  Google Scholar 

  49. Akillioglu HG, Karakaya S. Changes in total phenols, total flavonoids, and antioxidant activities of common beans and pinto beans after soaking, cooking, and in vitro digestion process. Food Sci Biotechnol. 2010;19(3):633–9.

    Article  CAS  Google Scholar 

  50. Long PL, Millard BJ, Joyner LP, Norton CC. A guide to laboratory techniques used in the study and diagnosis of avian coccidiosis. Folia Vet Lat. 1976;6(3):201–17.

    CAS  PubMed  Google Scholar 

  51. Duszynski DW, Wilber PG. Critical comment. A guideline for the preparation of species descriptions in the Eimeriidae. J Parasitol. 1997;83:333–6.

    Article  CAS  PubMed  Google Scholar 

  52. Schito ML, Barta JR, Chobotar B. Comparison of four murine Eimeria species in immunocompetent and immunodeficient mice. J Parasitol. 1996;82:255–62.

    Article  CAS  PubMed  Google Scholar 

  53. Dommels YE, Butts CA, Zhu S, Davy M, Martell S, Hedderle D, Barnett MP, McNabb WC, Roy NC. Characterization of intestinal inflammation and identification of related gene expression changes in mdr1a(–/–) mice. Genes Nutr. 2007;2:209–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tsakiris S, Schulpis KH, Marinou K, Behrakis P. Protective effect of l-cysteine and glutathione on the modulated suckling rat brain Na+, K+, -ATPase and Mg2+ -ATPase activities induced by the in vitro galactosaemia. Pharm Res. 2004;49:475–9.

    Article  CAS  Google Scholar 

  55. Paglia DE, Valentine WN. Studies on the Quantitative and Qualitative Characterization of Erythrocyte Glutathione Peroxidase. J Lab Clin Med. 1967;70:158–69.

  56. Nishikimi M, Roa NA, Yogi K. Biochem Bioph Res Common. 1972;46:849–54.

    Article  CAS  Google Scholar 

  57. Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurement of antioxidant activity in human fluids. J Clin Pathol. 2001;54:356–61.

  58. Dkhil MA, Abdel Moneim AE, Bauomy AA, Khalil M, Al-Shaebi EM, Al-Quraishy S. Chlorogenic acid prevents hepatotoxicity in arsenic-treated mice: role of oxidative stress and apoptosis. Mol Biol Rep. 2020;47:1161–71.

    Article  CAS  PubMed  Google Scholar 

  59. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2–∆∆CT method. Methods. 2001;25:402–8.

    Article  CAS  PubMed  Google Scholar 

  60. Ding H, Han C, Guo D, Chin YW, Kinghorn AD, D’Ambrosio SM. Chemopreventive characteristics of avocado fruit. Semin Cancer Biol. 2007;17:386–94.

    Article  PubMed  Google Scholar 

  61. Araújo RG, Rodriguez-Jasso RM, Ruiz HA, Pintado MME, Aguilar CN. Avocado by-products: nutritional and functional properties. Trends Food Sci Technol. 2018;80:51–60.

    Article  Google Scholar 

  62. Velderrain-Rodríquez GR, Quero J, Osada J, Martín-Belloso O, Rodríquez-Yoldi MJ. Phenolic-rich extracts from avocado fruit residues as functional food ingredients with antioxidant and antiproliferative properties. Biomolecules. 2021;11(7): 977.

    Article  Google Scholar 

  63. Vinha F, Moreira A, Barreira J. Physicochemical parameters, phytochemical composition and antioxidant activity of the algarvian avocado (Persea americana Mill). J Agric Sci. 2013;5(12):100–9.

    Google Scholar 

  64. -Chia TWR, Dykes GA. Antimicrobial activity of crude epicarp and seed extracts from mature avocado fruit (Persea americana) of three cultivars. Pharm Biol. 2010;48(7):753–6.

    Article  Google Scholar 

  65. Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev. 2013;26(1):58–85.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Abdel-Tawab H, Abdel-Baki AS, El-Mallah AM, Al-Quraishy S, Abdel-Haleem HM. In vivo and in vitro anticoccidial efficacy of Astragalus membranaceus against Eimeria papillata Infection. J King Saud Uni Sci. 2020;32(3):2269–75.

    Article  Google Scholar 

  67. Elmahallawy EK, Fehaid A, El-Shewehy DMM, Ramez AM, Alkhaldi AAM, Mady R, Nasr NE, Arafat N, Hassanen EAA, Alsharif KF, Abdo W. S-Methylcysteine ameliorates the intestinal damage induced by Eimeria tenella Infection via targeting oxidative stress and inflammatory modulators. Front Vet Sci. 2022;8: 754991.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Vo TS, Le PU, Ngo DH. Free radical scavenging and anti-proliferative activities of avocado (Persea americana Mill.) Seed extract. Asian Pac J Trop Biomed. 2019;9(3):91–7.

    Article  CAS  Google Scholar 

  69. Lüder CG, Gross U, Lopes MF. Intracellular protozoan parasites and apoptosis: diverse strategies to modulate parasite-host interactions. Trends Parasitol. 2001;17:480–6.

    Article  PubMed  Google Scholar 

  70. Balamurugan K, Rajaram R, Ramasami T. Caspase-3: its potential involvement in cr(III)-induced apoptosis of lymphocytes. Mol Cell Biochem. 2004;259:43–51.

    Article  CAS  PubMed  Google Scholar 

  71. Alkhudhayri AA, Dkhil MA, Al-Quraishy S. Nanoselenium prevents eimeriosis-induced inflammation and regulates mucin gene expression in mice jejunum. Int J Nanomed. 2018;13:1993.

    Article  CAS  Google Scholar 

  72. Bonilla-Porras AR, Salazar-Ospina A, Del Rio MJ, Pereañez-Jimenez A, Velez-Pardo C. Pro-apoptotic effect of Persea americana var. Hass (avocado) on Jurkat lymphoblastic Leukemia cells. Pharm Biol. 2013;52(4):458–65.

    Article  Google Scholar 

  73. Abozaid OAR, Anees LM, Abdel-Hamed GR. Modulatory effect of Persea americana oil against diethylnitrosamine-induced hepatotoxicity in rats: a proposed mechanism. Bull Natl Res Cent. 2021;45:193.

    Article  Google Scholar 

  74. El-Magd MA, Zedan AMG, Zidan NS, Sakran MI, Bahattab O, Oyouni AAA, Al-Amer OM, Alalawy AI, Elmoslemany AM. Avocado seeds-mediated alleviation of cyclosporine A-induced hepatotoxicity involves the inhibition of oxidative stress and proapoptotic endoplasmic reticulum stress. Molecules. 2022;27(22): 7859.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Knoop KA, Newberry RD. Goblet cells: multifaceted players in immunity at mucosal surfaces. Mucosal Immunol. 2018;11(6):1551–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Khan WI. Physiological changes in the gastrointestinal tract and host protective immunity: learning from the mouse-trichinella spiralis model. Parasitol. 2008;135:671–82.

    Article  CAS  Google Scholar 

  77. Forder RE, Nattrass GS, Geier MS, Hughes RJ, Hynd PI. Quantitative analyses of genes associated with mucin synthesis of broiler chickens with induced necrotic enteritis. Poult Sci. 2012;91(6):1335–41.

    Article  CAS  PubMed  Google Scholar 

  78. Alkhudhayri A, Al-Shaebi EM, Qasem MAA, Murshed M, Mares MM, Al-Quraishy S, Dkhil MA. Antioxidant and anti-apoptotic effects of selenium nanoparticles against murine eimeriosis. An Acad Bras Cienc. 2020;92(2): e20191107.

    Article  CAS  PubMed  Google Scholar 

  79. Alkhudhayri A, Thagfan FA, Al-Quraishy S, Abdel-Gaber R, Dkhil MA. Assessment of the oxidative status and goblet cell response during eimeriosis and after treatment of mice with magnesium oxide nanoparticles. Saudi J of Biol Sci. 2022;29:1234–8.

    Article  CAS  Google Scholar 

  80. Yunus M, Horil Y, Makimura S, Smith AL. Murine goblet cell hypoplasia during Eimeria pragensis Infection is ameliorated by clindamycin treatment. J Vet Med Sci. 2004;67(3):311–5.

    Article  Google Scholar 

  81. Tan J, Applegate TJ, Liu S, Guo Y, Eicher SD. Supplemental dietary L-arginie attenuates intestinal mucosal disruption during a coccidial vaccine challenge in broiler chickens. Br J Nutr. 2014;112(7):1098–109.

    Article  CAS  PubMed  Google Scholar 

  82. Kopp ZA, Jain U, van Limbergen J, Stadnyk AW. Do antimicrobial peptides and complement collaborate in the intestinal mucosa? Front Immunol. 2015;6:17.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by the Researchers Supporting Project (RSP2023R25), King Saud University, Riyadh, Saudi Arabia, and also supported by Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2023R437), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.A.-G. and S.A.Q.; methodology, R.A.-G., T.A.-O. and M.A.D; software, R.A.-G., E.M.A.-S. and M.A.D.; validation, M.A.D., F.A., and S.A.Q.; formal analysis, R.A.-G., E.M.A.-S. and N.A.-H.; investigation, R.A.-G. and M.A.D.; resources, R.A.-G. and M.A.D.; data curation, R.A.-G. and M.A.D.; writing—original draft preparation, R.A.-G., T.A.-O., and M.A.D.; writing—review and editing, M.A.D. and R.A.-G.; funding acquisition, R.A.-G. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Rewaida Abdel-Gaber.

Ethics declarations

Ethics approval and consent to participate

This research was approved by the Research Ethics Committee (REC) at King Saud University (approval number KSU-SE-22-40). All methods were carried out following relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alajmi, F., Al-Otaibi, T., Al-Quraishy, S. et al. Persea americana extract protects intestinal tissue from Eimeria papillata-induced murine Infection. BMC Vet Res 19, 248 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s12917-023-03810-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12917-023-03810-1

Keywords