Next Article in Journal
Acclimation Effects of Natural Daily Temperature Variation on Longevity, Fecundity, and Thermal Tolerance of the Diamondback Moth (Plutella xylostella)
Next Article in Special Issue
Basic Structures of Gut Bacterial Communities in Eusocial Insects
Previous Article in Journal
Larvicidal Activity of Carbon Black against the Yellow Fever Mosquito Aedes aegypti
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Functional Properties and Antimicrobial Activity from Lactic Acid Bacteria as Resources to Improve the Health and Welfare of Honey Bees

1
Department of Agricultural, Environmental and Food Sciences (DiAAA), University of Molise, Via De Sanctis snc, 86100 Campobasso, Italy
2
Conaproa, Consorzio Nazionale Produttori Apistici, 86100 Campobasso, Italy
*
Author to whom correspondence should be addressed.
Insects 2022, 13(3), 308; https://doi.org/10.3390/insects13030308
Submission received: 24 January 2022 / Revised: 17 March 2022 / Accepted: 19 March 2022 / Published: 21 March 2022
(This article belongs to the Special Issue Gut Microbiota in Social Insects)

Abstract

:

Simple Summary

Honey bees play a pivotal role in the sustainability of ecosystems and biodiversity. Many factors including parasites, pathogens, pesticide residues, forage losses, and poor nutrition have been proposed to explain honey bee colony losses. Lactic acid bacteria (LAB) are normal inhabitants of the gastrointestinal tract of honey bees and their role has been consistently reported in the literature. In recent years, there have been numerous scientific evidence that the intestinal microbiota plays an essential role in honey bee health. Management strategies, based on supplementation of the gut microbiota with probiotics, may be important to increase stress tolerance and disease resistance. In this review, recent scientific advances on the use of LABs as microbial supplements in the diet of honey bees are summarized and discussed.

Abstract

Honey bees (Apis mellifera) are agriculturally important pollinators. Over the past decades, significant losses of wild and domestic bees have been reported in many parts of the world. Several biotic and abiotic factors, such as change in land use over time, intensive land management, use of pesticides, climate change, beekeeper’s management practices, lack of forage (nectar and pollen), and infection by parasites and pathogens, negatively affect the honey bee’s well-being and survival. The gut microbiota is important for honey bee growth and development, immune function, protection against pathogen invasion; moreover, a well-balanced microbiota is fundamental to support honey bee health and vigor. In fact, the structure of the bee’s intestinal bacterial community can become an indicator of the honey bee’s health status. Lactic acid bacteria are normal inhabitants of the gastrointestinal tract of many insects, and their presence in the honey bee intestinal tract has been consistently reported in the literature. In the first section of this review, recent scientific advances in the use of LABs as probiotic supplements in the diet of honey bees are summarized and discussed. The second section discusses some of the mechanisms by which LABs carry out their antimicrobial activity against pathogens. Afterward, individual paragraphs are dedicated to Chalkbrood, American foulbrood, European foulbrood, Nosemosis, and Varroosis as well as to the potentiality of LABs for their biological control.

1. Introduction

Honey bees play a crucial role in the maintenance of wider biodiversity, ecosystem stability, and agricultural production through pollination [1,2,3]. While global stocks of managed honey bee colonies appear to be increasing, significant decline and colony losses of wild and domestic bees have been reported in many parts of the world [4,5,6,7].
Multiple biotic and abiotic factors are associated with the honey bee colony losses [4,8,9,10,11,12,13]. Furthermore, there is a growing consensus that parasites and pathogens are among the most significant threats to the management of bee colonies [14,15].
In-depth knowledge of these factors is essential and a prerequisite for developing measures to ensure both healthy bees and sustainable pollination. The aforementioned factors can also influence the honey bees’ gut microbiota. Its dysbiosis could weaken the honey bees, thus contributing to the phenomenon of Colony Collapse Disorder (CCD) [16,17,18]. Moreover, the gastrointestinal tract of honey bee’s larvae and adult bees is the infection and transmission site of different pathogens, such as Ascosphaera apis, Nosema ceranae, Paenibacillus larvae, Melissococcus plutonius, and viruses. These pathogens may cause economic losses in agriculture, affecting the survival of managed and wild honey bees [19,20,21,22]. Recently, many different control measures, such as fungicides, antibiotics, heterocyclic organic compounds (indoles), and bacteriophages, have been used to control honey bee diseases [23,24]. Most of these products were promising in terms of controlling the growth of pathogens both in vitro and in vivo. Nevertheless, these approaches could be useful as therapy, but are often ineffective for prophylactic purposes, leaving the honey bee colonies vulnerable to diseases. In addition, the use of antibiotics in beekeeping is legally banned in many countries of the European Union [25], due to the risks for both human and honey bee health [17,26], and to the uncontrolled spread of antimicrobial genes [27]. Therefore, there is a growing interest in new effective means of controlling disease and improvement in honey bee health, as well as providing benefits for agriculture by increasing yield and quality of crop production. The use of naturally occurring compounds for disease control could be an interesting approach that needs to be further investigated because the findings to date have not always been of biological relevance [28,29,30].
The gut microbiota is fundamental for honey bee’s growth and development, immune function, protection against pathogen invasion [17]. A well-balanced microbiota is essential to support honey bee health and vigor, moreover, the structure of the intestinal bacterial community can become an indicator of the honey bee health [31,32,33].
The gut microbial communities can also provide an important new tool to improve disease management strategies and contribute to the development of novel and sustainable disease monitoring approaches [34,35,36]. More in general, the manipulation and the exploitation of the insect microbiota could be effective in developing strategies for the management of insect-related problems [37,38]. Indeed, this approach, generally defined as ‘Microbial Resource Management’ (MRM), was described as ‘Symbiont Resource Management’ (SMR) when applied to symbiotic microorganisms. The MRM refers to the proper management of the microbial resource, present in a given ecosystem, in order to solve practical problems through the use of microorganisms. One of the environmental hot topics is represented by the gastrointestinal tract (GIT) defined as an “outside world within living animals” [39]. The main objective is to control and steer microbial communities, and microbial processes, in the most sustainable way.
Moreover, the protection against pathogens and/or parasites is one of the frequently associated aspects of a balanced intestinal microbiota. Indeed, it is widely known that the early stages of pathogens infection can be eased by any nutritional or environmental stress causing microbial dysbiosis. In several studies, it has been proven that, among the microbial symbionts associated with the honey bee, the lactic acid bacteria have a probiotic effect on bees by stimulating their immunity and helping them to overcome pathogen attacks [40,41,42,43]. Different mechanisms, among which the direct pathogen inhibition by the release of antimicrobial compounds, the stimulation of the immune system, and the competitive exclusion, mediated by the microbial symbionts, could be involved in the honey bee protection.
In this review, we briefly introduce the presence and the role of the LAB community in the honey bees’ gut microbiota and, subsequently, we discuss the utilization of LABs as probiotics and their potential for the biocontrol of some honey bee diseases.

2. Gut Microbiota of Honey Bee: Presence and Role of Lactic Acid Bacteria (LAB)

The GIT microbiota structure of honey bees (Apis mellifera) is both unique and highly specialized; in detail, the dominant bacterial phyla belong to Proteobacteria, Actinobacteria, Bacteroidetes, and Firmicutes [44,45]. In social insects, such as the honey bees, the intestinal bacteria are transmitted and shared by colony members through oral–fecal and trophallactic transmission. However, consumption of stored pollen or bee bread, contact with older bees within the hive, and hive material during the adult phase are also involved in transmitting and sharing the bacteria [17,44,45].
The lactic acid bacteria (LAB) are normal inhabitants of the GIT of many insects, and their presence in the honey bee intestinal tract has been consistently reported in the literature [46,47,48]. These bacteria belong to a biologically defined group where lactic acid is produced during homo- or hetero-fermentative metabolism.
Soil and plants are considered the hypothetical first niche of the ancestral LAB, followed by the gut of herbivorous animals [49]; the transition from the soil and the plants to the animal gut occurred by three areas of genomic adaptation [50,51]: resistance to host barriers, adhesion to intestinal cells, and fermentation.
A bees’ gut is the optimal microenvironment for LAB as it is defined by microaerobic conditions, presence of nectar and sugars, optimal temperature. Olofsson et al. [52] suggested that bees and their microbiota are mutually dependent, in fact, LABs received a niche in which nutrients were available and bees gained protection [35,53,54].
LABs’ importance is further emphasized by their ecological distribution, which is not limited to adult bee gut only, in fact, they have been isolated from larval guts [55] and the honey stomach of adult bees [56]. This latter structure, adjacent to the midgut, is a further relevant microbial niche associated with food storage and liquid transfer (water, nectar, and royal jelly). In addition, LABs are also dominant in the hive environment (beebread, honey, wax, and comb) [46,47,48,52,53,54,56,57,58].
LABs have also been extensively studied in animals and humans because of their probiotic properties, which have led to their well-built commercial exploitation in the food, feed, and pharmaceutical market [59,60,61,62,63]. The finding that a component of the honey bee gut microbiota was represented by LABs has increased the interest of scientists in looking for similarities and analogies with the probiotic bacteria widely investigated in humans and animals.

3. Functional Properties of LAB

There are several properties through which LABs can provide a specific health benefit for the honey bees [60,61,62].
The following section discusses some functional properties of LAB supplemented as probiotics in bee feeding. Table 1 provides an overview of the main results obtained in several studies.

3.1. Heavy Metals Detoxification

Global environmental pollution is increasing because of continuous and intense industrial and technological development, in particular, toxic metals, such as heavy metals (HM), are considered one of the major harmful pollutants [81,82,83,84]. HMs, once found in soil or water, are not easily eliminated; they cause irreversible damage to the survival, feeding, growth, and impair the behavior of the organisms, including honey bees [85,86,87,88,89]. It has been widely demonstrated that the honey bee populations are susceptible to several environmental threats, including HMs [81]. The honey bees can be exposed to HMs when foraging on contaminated honey and pollen resources and, in some cases, by airborne exposure [90,91]. The HMs also bioaccumulate in larval and adult stages, in the colony’s honey, wax, and propolis supplies [86,91], making the honey bees excellent bioindicators of HMs presence in the environment [92,93]. Some reports indicate that the honey bee cell ultrastructure can be adversely affected by HMs, inducing cell apoptosis that can disrupt cell vigor and cell proliferation. HMs can also negatively affect the genetic material, resulting in mutation, and in addition, they also cause neurotoxic effects [86,89,91,94,95,96,97,98,99,100]. Other studies have shown that the HMs may affect antioxidant capacity and immunocompetence in honey bees [87,88,101].
Many bacteria, including the LABs, appear to have the ability to efficiently remove the HMs through two mechanisms: biosorption and bioaccumulation [102,103,104,105].
Biosorption refers to the binding of metals onto the cell wall’s surface and it is a simple physicochemical process, whereas the bioaccumulation process refers to the intracellular accumulation of metals that occur in two stages, biosorption and bioaccumulation by transporting the metals across the cell wall and membrane [106,107,108]. Recently, the next generation of probiotics has attracted increasing attention [103,109,110] for their ability to alleviate HMs toxicity, although, most of the studies have been performed with an in vitro digestion or animal model. [75,103,111,112,113,114,115,116,117,118]. Based on this research, specific LABs could be used as a new dietary therapeutic strategy against HMs toxicity. In this regard, Rothman et al. [85] demonstrated that some honey bee symbiotic LABs are capable of in vitro metals’ bioaccumulation. However, these results are preliminary and so, more in-depth, systematic, and epidemiological studies need to be performed on honey bees.

3.2. Mitigation of Pesticides Effect

Pesticides, such as insecticides and fungicides, are considered one of the possible stressors causing the general decline in honey bees and colony losses [5,12,119]. The exposure of honey bees to pesticides also causes microbial dysbiosis and immunosuppression, rendering them more susceptible to pathogens; furthermore, the interactions between pesticides and pathogens may exacerbate honey bees’ mortality [120,121,122,123,124,125,126,127,128,129,130]. A novel concept may be the administration of lactic acid bacteria to mitigate the harmful effects of pesticides. There are several mechanisms through which the treatment with probiotics could act on pesticide intoxication; for example, the treatment with Pediococcus acidilactici restored the expression of two genes, which were altered by pesticide co-exposure, coding for serine protease 40 and vitellogenin [131]. Moreover, the benefit of LAB supplementation is a reduction in pesticide uptake through their degradation [132,133,134,135] or sequestration of ingested organophosphate pesticides, which has been associated with reduced intestinal absorption and insect toxicity with appropriate models [136,137]. In other model organisms, LABs have been shown to reduce toxicity and exert a protective effect on the host [135,138,139,140], thus establishing a basis for future studies to investigate this potential in honey bees. Recently, some authors have highlighted how the resistance and capacity of LABs for degrading organophosphorus pesticides is strain-dependent [134,141] and showed the feasibility of the LAB to be developed into probiotic products capable of alleviating oxidative damage caused by pesticides in vivo [142].
Based on this knowledge, probiotic supplementation with appropriate LAB cultures could mitigate the sublethal effects of pesticides by reducing pesticide uptake, improving pathogen resistance, and mitigating sublethal effects on colony development. Until chemical agents are no longer used in agriculture, the ability to supplement honey bees with probiotics could help the insects to fight the unintended pernicious effects [143].

3.3. Adhesion to Intestinal Mucosa and Enhancement of the Epithelial Barrier

Adhesion to intestinal epithelial cells is a prerequisite for the colonization of probiotic bacteria, leading to transitory colonization that would foster the immune response and, at the same time, stimulate the intestinal barrier and metabolic functions. In addition, this ability to adhere to the host may serve a protective role against undesirable microorganisms through competition for host cell-binding sites [108,144,145,146].
As reported in a number of studies, during this adaptation phase, bacteria produce extracellular polymeric substances (EPS), containing biological macromolecules, some of which (polysaccharides, proteins, nucleic acids, and lipids) are also responsible for the cohesion of microorganisms and are implicated in the production of biofilms [146,147].

3.4. Participation in the Digestive Process

The honey bee gut microbiota, as well as that of other insects, synthesize essential nutritional compounds and improve the digestion efficiency and availability of nutrients [17,47,148]. A properly functioning gut microbiota is closely connected to the health of the honey bee since it provides countless enzymatic activities to break down the complex sugars of the honey bees’ diet. Iorizzo et al. [48] proved that some Lactiplantibacillus plantarum (previously Lactobacillus plantarum) strains isolated from honey bee gut possess both alpha- and beta-glycosidase activities. The enzyme beta-glycosidase in association with other enzymes, cellulase, and hemicellulase produced by bee intestinal symbionts, such as Gilliamella, contributes to the hydrolysis of cellulose [149]. The alpha-glycosidase converts maltose to glucose and with alpha-amylase, is involved in the starch breakdown [150].
Honey bees collect food rich in carbohydrates, such as sucrose, glucose, and fructose, which are important for the development and well-being of their colonies [151]. However, other carbohydrates present in their diet in lesser quantities, such as monosaccharides (e.g., mannose, galactose, xylose, arabinose, rhamnose) and oligosaccharides (e.g., lactose, melibiose, raffinose, and melezitose), may be toxic to bees as they do not have specific enzymatic activity for their metabolization [152,153]. Iorizzo et al. [154,155] evidenced that some Apilactibacillus kunkeei (previously Lactobacillus kunkeei) and Lp. plantarum strains can metabolize arabinose, galactose, lactose, mannose, melibiose, melezitose, and raffinose. As they are able to simultaneously intervene in the breakdown of complex polysaccharides and metabolize toxic sugars, the role of LABs in enhancing food tolerance and maintaining the health of their hosts could be considerable [156].

3.5. Antioxidative Activity

Recent research demonstrated that several biotic and abiotic factors, induce oxidative stress and impair the antioxidant defensive capacity of honey bee larvae [9,157,158,159,160].
Oxidative stress is an important process that can cause severe negative effects in eukaryotic organisms. Reactive oxygen species (ROS) are produced during normal metabolic processes and are responsible for oxidative stress. To prevent or reduce ROS-induced oxidative stress, insects use various enzymatic mechanisms that cause oxidative inactivation (superoxide dismutase, catalase, and peroxidase) or removal of ROS at the intracellular level through the enzymes glutathione peroxidase (GPX) and glutathione reductase (GSR) [161,162,163].
These particular enzyme activities are relevant for the health of honey bees when they are under biotic and abiotic stressors, such as nutritional and thermal stress, parasites, heavy metals, and/or pesticides [9,129,157,164,165,166,167,168]. Oxidative stress can also be a consequence of some honey bee diseases; in fact, during the excessive growth of pathogens, the levels of ROS in the infection site increase [160]. Dussaubat et al. [169], and more recently Li et al. [158], reported that the oxidative stress in honey bee larvae and the decreased levels of metabolites involved in mitigating oxidative stress induced by Ascosphera apis could disrupt the antioxidant defenses of the infected larvae. Antioxidant enzymatic activity and the amounts of certain metabolites (e.g., taurine, docosahexaenoic acid, and L-carnitine) involved in reducing oxidative stress were significantly decreased in the gut of infected larvae [158]. In recent years, particular attention has been focused on the application of LABs as natural antioxidants. Some strains belonging to this group have both enzymatic and non-enzymatic antioxidant activity, which can reduce the oxidative damage caused by the accumulation of ROS during the digestive process [170,171,172]. Probiotic LABs have complex antioxidant mechanisms, and different strains use different mechanisms: chelation of toxic ions (Fe2+ and Cu2+); synthesis of antioxidant compounds (e.g., glutathione, butyrate, folate, and exopolysaccharides); activation of transcription of enzymes that neutralize free radicals [173,174,175]. Further research aimed at the selection, and diet utilization, of appropriate probiotics that can contribute to the reduction in oxidative stress in honey bees, would be interesting.

4. Antimicrobial Activity of LAB

The first part of this section discusses some of the mechanisms by which LABs carry out their antimicrobial activity against pathogens. Afterward, individual paragraphs are dedicated to some main diseases and the potentiality of LABs for their biological control. Table 2 provides an overview of the main results obtained in recent years.

4.1. Immune System Stimulation

Several abiotic and biotic factors, which include pathogens (parasites, fungi, bacteria, and viruses), the alteration or loss of ecosystems, and/or the use of agrochemicals, are contributing to the decline in bee populations. All these factors alter the defense mechanisms of the bee immune system [33,140,203,204,205,206,207] which includes physical barriers and generalized cellular and humoral responses to infectious and parasitic organisms [208,209]. The humoral response is the most important defense system in the honey bee and is mediated by the production of antimicrobial peptides (AMPs), such as apidaecin, abaecin, hymenoptaecin, and defensin [210]. These compounds act by restricting the enzymes necessary for pathogen growth and duplication, forming channels in the pathogens’ cell membranes [211]. Probiotic bacteria can exert an immunomodulatory effect, in particular Lactobacilli and Bifidobacteria, but also the gamma-proteobacterium Frischella perrara have shown to enhance the honey bee immune system [76,212,213]. Yoshiyama et al. [80], using a mixture of probiotic bacteria strains, (one strain of Enterococcus thailandicus, three of Weissella cibaria, one of W. viridescens, and two of Lactobacillus curvatus), achieved a significant upregulation in abaecin, defensin, and hymenoptaecin peptides, both in adult and larvae bees. The combination of Bifidobacterium longum, B. breve, B. infantis, Lactobacillus rhamnosus, L. reuteri, L. acidophilus, Lp. plantarum, and L. casei caused the AMPs upregulation, which boosted the honey bee immune system [76]. In addition, B. asteroides and Fructobacillus pseudoficulneus strains, isolated from honey bee worker gut and bee bread, exerted a considerable upregulation in apidaecin synthesis, and the effects on the honey bee immune system are strain-specific [78]. Moreover, a pollen-based probiotic preparation improved immunity in bee colonies, and thus increased their resistance to infectious diseases and stressful conditions [74].

4.2. Competitive Exclusion of Pathogenic Microorganisms

Many honey bee pathogens need to adhere to the mucosal layer of the gastrointestinal tract to start the infectious process. By colonizing the intestine and adhering to its epithelial surface, bacteria interfere with the adhesion of pathogens. [154]. This property is also known as “competitive exclusion” [214]. The existence of any microbial population depends on its ability to compete for chemicals and available energy with other microbes in the same environment. Some probiotic LABs consume nutrients or sequester chemicals that are essential for the growth of pathogens. For example, siderophores are low-molecular-weight ferric iron-chelating agents that are able to dissolve precipitated iron, or extract it from iron complexes, to make it available for bacterial growth [214,215].

4.3. Production of Antimicrobial Substances

Probiotic bacteria produce bactericidal or bacteriostatic substances, including bacteriocins, hydrogen peroxide, siderophores, lysozymes, proteases, which act on other microbial populations [214,216,217,218,219,220]. In addition, some bacteria produce organic acids, and volatile fatty acids (e.g., lactic, acetic, butyric, and propionic acids), that can result in the reduction in pH in the gastrointestinal lumen, thus preventing the growth of opportunistic pathogenic microorganisms [61,216]. The processes behind the inhibition activity may include a number of compounds that can lead to membrane disruption (fatty acids or peptides), H+ gradient interference, or enzyme inhibition. In addition, the LAB antimicrobial action is often due to a complex interaction among different compounds [155,217]. Organic acids, particularly acetic acid and lactic acid, have a strong inhibitory effect against Gram-negative bacteria, and they have been considered the main antimicrobial compounds responsible for the probiotic inhibitory activity against pathogens. The undissociated form of the organic acid enters the bacterial cell and dissociates inside the cytoplasm. The eventual lowering of the intracellular pH or the intracellular accumulation of the organic acid ionized form can lead to pathogen death [218]. In addition, many LABs produce antibacterial peptides, including bacteriocins and small AMPs, whose common mechanisms of bacteriocin-mediated killing include the destruction of target cells by pore formation and/or inhibition of cell wall synthesis [219]. Bacteriocins exhibit antimicrobial activity with a variable spectrum depending on the peptide, which may target several bacteria [221,222,223]. Moreover, there is scientific evidence that bacteriocins are effective against some bacterial infections of honey bees [224]. Some studies, which will be cited later, conducted on the antimicrobial activity of probiotic LAB (Table 2), have not shown biological relevance due to the partial inhibition of pathogens. However, it should be noted that the benefit of probiotics is the result of a sum of actions (antimicrobial, detoxifiers, antioxidants, and immunostimulants) that overall can improve the overall health and well-being of bees.

4.4. Chalkbrood

Chalkbrood is a fungal honey bee brood disease caused by Ascosphaera apis [225,226], a heterothallic parasitic fungus, belonging to the phylum Ascomycota. This disease is currently widespread throughout the world and different studies have pointed out that chalkbrood incidence may be increasing [225,227]. This economically important disease, although it does not destroy the entire colony, weakens it, and can lead to significant losses of both honey bees and colony productivity [228,229,230]. In addition, A. apis infection, inducing oxidative stress in honey bee larvae, may compromise their antioxidant defensive capacity [158].
Over the years, several chemotherapeutic compounds and strategies have been tested and developed to control the chalkbrood disease [231,232,233,234,235,236], but to date, there are no effective compounds to properly control the disease. Moreover, chemotherapeutic compounds are often expensive and release residues in honey bee products, representing a risk for human health [237], as well as causing an imbalance in the normal honey bee intestinal microbiota [238]. Therefore, a growing interest has been reported in developing chalkbrood-controlling strategies. Among natural compounds, several essential oils demonstrated a significant antifungal activity [239,240,241,242]. Propolis exhibits inhibiting activities against pathogens, including A. apis, and in particular, it has been observed that the effects depend on the plant resources of each region from which the propolis is produced [243,244,245,246].
In recent years, various studies have also led to the hypothesis that the honey bee intestinal microbial community may represent a future alternative strategy for the control of the chalkbrood disease. Bacillus subtilis, B. megaterium, and B. circulans [247] have shown in vitro inhibition activity against A. apis. Sabaté et al. [248] in a study on the inhibition of P. larvae and A. apis by Bacillus subtilis, hypothesized that B. subtilis secreted antimycotic compounds, and more recently Omar et al. [249] demonstrated an antagonistic activity of B. subtilis and Pseudomonas fluorescens strains, isolated from the gut of Apis mellifera carnica, against A. apis. Tejerina et al. [176] demonstrated that the in vitro application of L. melliventris LSAM, L. helsingborgensis LSAI, and A. kunkeei LSAJ strains, isolated from beebread, affected the growth and sporulation of A. apis. The same authors have shown that feeding honey bee larvae with LAB (L. melliventris, L. helsingborgensis, and A. kunkeei) in sugar syrup over 5 months, reduced larval mummification in chalkbrood disease by over 80% [176]. Iorizzo et al. [154,178] proved that L. kunkeei K7, K18, K34, K40, K41, K45, K55, K64, and K112 (current name Apilactobacillus kunkeei) and Lactiplantibacillus plantarum P8, P25, P86, P95, and P100 not only inhibited A. apis but were also suitable in the preparation of a “probiotic syrup” to reinstate the symbiotic communities of the honey bee intestine in case of dysbiosis and to perform a prophylactic action against A. apis.

4.5. American Foulbrood (AFB) and European Foulbrood (EFB)

Paenibacillus larvae (Bacillales, Paenibacillaceae), a flagellated, Gram-positive bacterium, is the causative agent of the quarantine disease American foulbrood (AFB), the most severe and cosmopolitan brood disease affecting A. mellifera larvae and pupae [14,15,21]. The infection follows the ingestion of spore-contaminated food provided to the larvae by honey bee nurses [250]. The spores germinate in the midgut lumen, where the vegetative bacteria massively proliferate, and secrete secondary metabolites, to counter microbial competitors, a chitin-degrading enzyme, responsible for the peritrophic matrix degradation [251,252,253], and other enzymes that allow the breaching of the midgut epithelium. The vegetative bacteria invade the hemocoel [254], causing the death of the larvae several days after infection, turning them into a ropy mass that dries and becomes a continuous source of infection, releasing a large number of bacterial spores [250]. These spores infect and kill a great number of larvae, resulting in the lack of offspring that leads to the collapse of the entire colony, causing considerable economic loss to beekeepers [21,255]. The infectious spores can spread within the colonies by nurse bees performing inhive tasks, such as cleaning, and through the feeding of larvae with spore-contaminated food [256]. The spore transmission can also occur between colonies through swarming, robbing, and drifting bees, as well as by beekeeping practices, e.g., through the movement of contaminated hive materials, such as honey or equipment [257,258,259]. P. larvae can produce very resilient spores which can remain viable for decades [260]. Melissococcus plutonius (Lactobacillales, Enterococcaceae) is a Gram-positive bacterium [261] that causes epidemic outbreaks of European foulbrood (EFB), one of the most worldwide detrimental brood diseases affecting honey bees, such as Apis mellifera L. [20,262]. Several other bacteria, typically found in the hive environment, may be associated with EFB disease, although the role of the secondary invaders in disease development is not yet entirely clear [20,55,263]. Honey bee larvae can be infected by ingestion of contaminated food, administered by adults [20], and once M. plutonius reach the intestinal tract, it quickly multiplies and can deprive the host of nutrients [264]. Diseased larvae are characterized by a typical change from white to a yellowish color, a foul or sour smell, they become flaccid, usually do not reach or complete the pupation stage and die 4–5 days after infection [20,265]. However, some infected larvae can survive and deposit the bacteria along with their feces in the comb, and they can survive long periods inside the hive [266]. M. plutonius in diseased colonies can be easily spread between colonies/apiaries [267,268].
The severe losses of brood, colony collapse, and extreme contagiousness make EFB and AFB economically important diseases, notifiable in many countries [15,269]. In most European countries, at least, the only measure to counteract them is the destruction of symptomatic colonies and the monitoring of neighboring apiaries to avoid pathogen spread [14,20,270]. However, strategies to treat and control the diseases vary across the world. In several countries, including the U.S. and Canada, antibiotics are commonly used for the treatment of infected colonies [15,271,272].
The use of antibiotics to control both diseases is an unsustainable strategy since they only affect vegetative form. The long-term treatment and their common use in prophylaxis, besides not eliminating the bacterial spores, has led to the development of antibiotic resistance [272,273,274,275,276]. Antibiotics also cause honey bee microbiota dysregulation [26], compromising the honey bee’s overall health status [17,18,31,268,277,278]; moreover, they may leave residues in honey [279,280]. For all these reasons, antibiotics are now strictly regulated or restricted in most European countries [279,280,281]. A recent study has investigated the potential of insect antimicrobial peptides for the development of a new class of insect-derived antibiotics to overcome AFB resistance to conventional antibiotics. The data obtained revealed a strong immune response against P. larvae in P. larvae-injected third instars of A. m. jemenitica and the presence of immune peptides in their plasma. Furthermore, the immune peptide fractions exerted significant in vivo therapeutic effects on P. larvae-infected first instars [282].
The application of alternative, natural strategies for EFB and AFB control could represent a suitable management approach [250,283]. Plant and propolis extracts, herbs, spices, and essential oils exhibit antimicrobial activity against P. larvae and M. plutonius, but, most of them have only been tested in vitro [250,284,285,286,287,288,289,290]. Macelignan and corosolic acid, the latter extracted from the banaba (Lagerstroemia speciosa) leaf, showed strong in vitro antibacterial activity against both P. larvae and M. plutonius [289]. Several studies are showing the in vitro ability of propolis extracts, from different botanical origins, or their constituents, to inhibit the growth of P. larvae [245,284,291,292,293]. Borba and Spivak [294], using colonies with and without a propolis envelope, demonstrated that the presence of the envelope provides an antimicrobial layer around the colony that protects the brood from P. larvae infection, resulting in a lower colony-level infection load. Some essential oils were tested in in vitro assays against P. larvae and M. plutonius [240,286,287,290,295,296]. The systemic administration of Cinnamomum zeylanicum EO to artificially infected nuclei of honey bee colonies prevented and controlled AFB [297,298]. The utilization of a favorable bacteria-based strategy for the prevention and biocontrol of honey bee pathogenic microorganisms offers interesting perspectives [42,43,131]. Different isolates of the bacteriocin-producing Enterococcus, often isolated from freshly collected pollen granules, showed strong in vitro inhibitory activity against P. larvae [188,189,190,191]. Yoshiyama et al. [80] demonstrated that nine LAB strains, belonging to Enterococcus sp., Weissella sp., and Lactobacillus sp., showed strong in vitro antagonistic activity against P. larvae, soon after oral administration, they were able to stimulate the honey bee innate immune response in vivo, which may be useful for preventing bacterial diseases in honey bees. A number of investigations also demonstrated that different strains belonging to LABs, among them A. kunkeei and Lp. plantarum, could be useful against P. larvae, inhibiting its survival and reducing pathogen load [177,180,181,183,185,186,187]. More recently, Iorizzo et al. [155] demonstrated that Lactiplantibacillus plantarum P8, P25, P86, P95, and P100 exhibited in vitro antagonistic activity against P. larvae, showing, at the same time, suitable physical and biochemical characteristics for their use as probiotics in the honey bee diet. In vivo studies demonstrated that the addition of gut bacteria to the diet significantly reduced the mortality of the infected A. m. jemenitica larvae [209] and that a diet supplemented with lactobacilli improved A. mellifera survival and hive resilience against P. larvae [214].
An in vitro study by Vasquez et al. [192] reported that A. kunkeei FF30-6, isolated from honey bees, showed antagonistic effects against M. plutonius, and this activity may be due to the production of anti-M. plutonius peptides or proteins [46,193]. Killer et al. [182] proved that Lactobacillus apis R4B, isolated from the stomach of honey bees, exhibits in vitro antagonistic effects against both P. larvae and M. plutonius. Another recent study has shown that the A. kunkeei V18 strain exhibit a strong in vitro inhibition for both P. larvae and M. plutonius [177]. Bacteria isolated from the gut of Apis cerana japonica F. have been shown to inhibit in vitro the growth of P. larvae [299]; while in vivo feeding bioassays proved that an isolate belonging to the genus Bacillus exhibited inhibitory activity against M. plutonius [36].

4.6. Nosemosis

Nosema apis (Zander 1909) and N. ceranae, [300] belonging to Microsporidia, obligate intracellular and spore-forming parasites, classified as fungi [301,302]. Both these species are etiologic agents of nosemosis, one of the most widespread and serious diseases of the adult honey bee [22,303,304,305]. N. apis is responsible for nosemosis type A [306], a disease that increases bee mortality in winter and causes a slow build-up in spring, makes weak and crawling bees, and reduces honey yield [307]. The disease also altered the infected worker bees’ flight behavior, reducing foraging ranges of colonies, and compromising their ability to provide pollination services [308]. N. ceranae provokes the nosemosis type C [306] that includes a wide range of effects on honey bees’ physiology and behavior, causes weakness, increases in colony mortality, and decreases in honey production; all factors that can contribute to colony collapse [309,310,311,312,313,314,315,316]. Moreover, N. ceranae infection can affect the host’s immune response leading to immune system suppression [204,206]. The acquisition of Nosema occurs via the fecal–oral pathway through the spores passed out of the host in excrement, which can contaminate the nesting environment, comb, floral resources, collected pollen, and water [22,307,310,317].
Because of the serious consequences of Nosema infections, there is a strong demand for the management of these pathogens. The antifungal fumagillin, used against nosemosis for several decades, has been banned by many countries (including the European Union) [318] due to its genotoxic and tumorigenic properties towards humans and toxicity to bees [319,320]. Therefore, the identification of alternative treatments is fundamental for honey bee health. Currently, several compounds, such as formic acid, are used for the management of Nosema infections [321]. Thymol and resveratrol were also reported to inhibit N. cerane [322,323,324]. Oxalic acid and Api-Bioxal®, a formulation based on dihydrate oxalic acid, were demonstrated to be active against N. ceranae, both in the laboratory and field [325,326,327]. The biological activity of other natural compounds towards Nosema infections has also been extensively explored; among them, essential oils and other organic extracts were reported to have anti-Nosema activity [328,329,330,331,332,333].
It is well known that midgut microorganisms have a part to play in the health of honeybees, and dietary integration with prebiotics and probiotics may offer a way to counteract Nosema infections. However, studies revealed that the administration of prebiotics or probiotics to honey bees has provided contrasting results. Chitosan, a prebiotic, increased honey bee resistance to N. apis infections [334], reduced N. ceranae spore load, and increased survivorship of the infected bees [335]. The administration of Brassicaceae defatted seed meals, as the integration of infected A. mellifera workers diet, leads to the inhibition of N. ceranae, and potential nutraceutical benefits were involved in the bee lifespan [336,337]. Some nutraceuticals, such as naringenin and carvacrol, and immuno-stimulatory compounds, administered in sugar syrup, reduced both spore multiplication and mortality during N. ceranae infection in honey bees [338]. Microbial supplements of the honey bee diet may have positive impacts on bee health and control nosemosis. Feeding the honey bee with the endogenous gut bacteria Bacillus subtilis reduced Nosema disease [248], and a surfactin, synthesized by the bacterium, was also proved to reduce N. ceranae development, acting either by direct exposure to purified spores or incorporated into the digestive tract of the bee [196]. Oral administration of organic acids, produced by L. johnsonii CRL1647, supplemented in syrup [67], provoked a Nosema ceranae occurrence reduction [66]. Parasaccharibacter apium improves honey bee resistance to Nosema, as well as the honey bee gut bacterial strains P. apium (PC1 sp.) and Bacillus sp. (PC2 sp.); moreover, the commercial probiotics Bactocell® R and Levucell SB® have increased the survival of Nosema infected bees. However, these last four probiotics do not exert a direct antagonistic effect on N. cerane development [200,339]. A Pediococcus acidilactici strain (PA CNCM MA18/5 M) was found to be the most efficient probiotic against N. ceranae, significantly improving the infected honey bee lifespan [131]. The commercial probiotic Vetafarm (L. acidophilus, L. delbruekii sub. bulgaricus, Lp. plantarum, L. rhamnosus, Bifidobacterium bifidum, Enterococcus faecium), besides reducing N. ceranae proliferation, increases the survival of infected honey bees [198]. Conversely, the bacteriocins by E. faecium CRL1385 and E. avium DSMZ 17,511 did not exert any activity towards N. ceranae [196]. Al. kunkeei and L. salivarius A3iob administered to honey bee colonies reduced Nosema disease [77]. Gajger et al. [77] proved that administering the commercial probiotic EM® PROBIOTIC FOR BEES was followed by a significant reduction in Nosema spp. spore counts in colonies and colonies’ strength were increased. On the contrary, the supplementation of the honey bees’ diet with the probiotic Lactobacillus rhamnosus and the prebiotic inulin showed no beneficial effect on the survival rates of honey bees infected with N. ceranae [199]. A significant increase in Nosema spore counts, compared to the A. mellifera infected control, was caused by a probiotic composed of L. casei, Lp. plantarum, Saccharomyces cerevisiae, and Rhodopseudomonas palustris [197,340]. RNA interference technologies, applied to both Nosema and honey bee, are also explored for Nosema disease control [301,302,303,304,305].

4.7. Varroosis

Varroa destructor is the main mite that can attack the honey bee (Apis mellifera L.). This parasite nourishes on the hemolymph of the honey bee and within a few years causes the colony to collapse [341,342]. Varroa mites can also host many pathogens, viruses, including Deformed wing virus (DWV) [343,344], and harmful bacteria [345]. Although in recent studies Ramsey et al. [342,346] proposed that V. destructor feeds on bee fat body, the caused damages always involve a reduction in protein content and hemocytes [347], leading to an imbalanced gut microbiota [348,349], and a decrease in host’s immune response [207,350,351]. Acaricides, such as fluvalinate, flumethrin, and amitraz, have successfully been used to control varroosis [352]. The use of these chemical compounds, however, has resulted in the development of resistant mites and the risk of acaricide residues in bee products, such as wax and honey. [353]. Therefore, there is an urgent need to find eco-friendly strategies that are both affordable and safe for humans and honey bees to control Varroa mites. Numerous studies have been conducted on natural remedies, such as plant extracts and essential oils, against V. destructor in honey bee colonies [354,355,356]. The efficacy of certain plant-based essential oils, such as eucalyptus, thyme, neem, sage, and grapefruit, against Varroa mites has been reported in many studies. [357,358,359,360]. However, the effectiveness of these methods is not constant and always effective, and the search for new strategies in Varroosis biocontrol is still ongoing [30,361]. Recent studies have highlighted the potential of LABs in the biocontrol of this disease. Tejerina et al. [194] demonstrated that L. salivarius A3iob reduced the levels of in situ varroosis by between 50 and 80%. L. johnsonii AJ5 and E. faecium SM21 strains enhanced bee survival and increased bee proteins [201]. Sacca and Lodesani [202] demonstrated in vitro that bacterial cultures of Al. kunkeei BO-G12 caused 95–100% mortality of the mite in 3 days.

5. Conclusions

A variety of adverse factors, among which pests and pathogens are the major candidates, but also pesticide exposure, diet quantity, quality, and diversity as well as unfavorable weather and fodder conditions, contribute to the decline in honey bees. In addition, the beekeeper’s management practices strongly influence the health of honey bee colonies. All these factors, acting alone or in combination, affect honey bee colonies, causing possible serious disturbances in the composition of the honey bee microbiota.
Moreover, honey bees, like all living things, are constantly in contact with many microorganisms that can exert an impact on their health and well-being.
Several researchers have investigated the use of the gut microbial symbionts as a supplement in the honey bee diet and the results have shown that such microorganisms (multiple strains or single strain), or their metabolites, could improve the health status of the honey bees. The honey bee symbionts could be exploited to actively counteract bee pathogens and parasites, or to enhance immunity, and thus indirectly, increase the protection of the honey bees’ health. The modulation of the honey bee gut microbiota, by supplementation of selected LABs, has aroused special attention since it represents a strategy to improve the health status of colonies, in terms of productivity, as well as boosting the presence of beneficial microorganisms within the gut of new-generation bees.
Moreover, appropriate probiotics could be exploited to actively counteract bee pathogens and parasites, or to enhance immunity, and thus indirectly, increase the protection of the honey bees’ health.
However, further studies are necessary to gain a better mechanistic understanding of how host–microbe–environment interactions influence honey bee mortality and colony loss.

Author Contributions

Conceptualization, M.I. and A.D.C.; software, F.L., B.T. and G.A.; writ-ing—original draft preparation, M.I., S.G., D.D.C. and S.P.; writing—review and editing, M.I., F.L. and G.A.; visualization, B.T.; supervision, M.I. and A.D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Potts, S.G.; Biesmeijer, J.C.; Kremen, C.; Neumann, P.; Schweiger, O.; Kunin, W.E. Global pollinator declines: Trends, impacts and drivers. Trends Ecol. Evol. 2010, 25, 345–353. [Google Scholar] [CrossRef] [PubMed]
  2. Klein, A.-M.; Vaissiere, B.E.; Cane, J.H.; Steffan-Dewenter, I.; Cunningham, S.A.; Kremen, C.; Tscharntke, T. Importance of pollinators in changing landscapes for world crops. Proc. R. Soc. B Biol. Sci. 2007, 274, 303–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Potts, S.G.; Imperatriz-Fonseca, V.; Ngo, H.T.; Aizen, M.A.; Biesmeijer, J.C.; Breeze, T.D.; Dicks, L.V.; Garibaldi, L.A.; Hill, R.; Settele, J. Safeguarding pollinators and their values to human well-being. Nature 2016, 540, 220–229. [Google Scholar] [CrossRef]
  4. Gray, A.; Brodschneider, R.; Adjlane, N.; Ballis, A.; Brusbardis, V.; Charriere, J.-D.; Chlebo, R.; Coffey, M.F.; Cornelissen, B.; Amaro da Costa, C. Loss rates of honey bee colonies during winter 2017/18 in 36 countries participating in the COLOSS survey, including effects of forage sources. J. Apic. Res. 2019, 58, 479–485. [Google Scholar] [CrossRef] [Green Version]
  5. Steinhauer, N.; Kulhanek, K.; Antúnez, K.; Human, H.; Chantawannakul, P.; Chauzat, M.-P. Drivers of colony losses. Curr. Opin. Insect Sci. 2018, 26, 142–148. [Google Scholar] [CrossRef] [PubMed]
  6. Moritz, R.F.; Erler, S. Lost colonies found in a data mine: Global honey trade but not pests or pesticides as a major cause of regional honeybee colony declines. Agric. Ecosyst. Environ. 2016, 216, 44–50. [Google Scholar] [CrossRef]
  7. Meeus, I.; Pisman, M.; Smagghe, G.; Piot, N. Interaction effects of different drivers of wild bee decline and their influence on host–pathogen dynamics. Curr. Opin. Insect Sci. 2018, 26, 136–141. [Google Scholar] [CrossRef] [PubMed]
  8. Alaux, C.; Le Conte, Y.; Decourtye, A. Pitting wild bees against managed honey bees in their native range, a losing strategy for the conservation of honey bee biodiversity. Front. Ecol. Evol. 2019, 7, 60. [Google Scholar] [CrossRef] [Green Version]
  9. Morimoto, T.; Kojima, Y.; Toki, T.; Komeda, Y.; Yoshiyama, M.; Kimura, K.; Nirasawa, K.; Kadowaki, T. The habitat disruption induces immune-suppression and oxidative stress in honey bees. Ecol. Evol. 2011, 1, 201–217. [Google Scholar] [CrossRef] [PubMed]
  10. Goulson, D.; Nicholls, E.; Botías, C.; Rotheray, E.L. Bee declines driven by combined stress from parasites, pesticides, and lack of flowers. Science 2015, 347, 1255957. [Google Scholar] [CrossRef]
  11. Otto, C.R.; Roth, C.L.; Carlson, B.L.; Smart, M.D. Land-use change reduces habitat suitability for supporting managed honey bee colonies in the northern great plains. Proc. Natl. Acad. Sci. USA 2016, 113, 10430–10435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Stanimirović, Z.; Glavinić, U.; Ristanić, M.; Aleksić, N.; Jovanović, N.; Vejnović, B.; Stevanović, J. Looking for the causes of and solutions to the issue of honey bee colony losses. Acta Vet. 2019, 69, 1–31. [Google Scholar] [CrossRef] [Green Version]
  13. Havard, T.; Laurent, M.; Chauzat, M.-P. Impact of stressors on honey bees (Apis mellifera; Hymenoptera: Apidae): Some guidance for research emerge from a meta-analysis. Diversity 2020, 12, 7. [Google Scholar] [CrossRef] [Green Version]
  14. Genersch, E. American Foulbrood in honeybees and its causative agent, PaeniBacillus larvae. J. Invertebr. Pathol. 2010, 103, S10–S19. [Google Scholar] [CrossRef] [PubMed]
  15. Genersch, E. Honey bee pathology: Current threats to honey bees and beekeeping. Appl. Microbiol. Biotechnol. 2010, 87, 87–97. [Google Scholar] [CrossRef]
  16. Cox-Foster, D.L.; Conlan, S.; Holmes, E.C.; Palacios, G.; Evans, J.D.; Moran, N.A.; Quan, P.-L.; Briese, T.; Hornig, M.; Geiser, D.M. A Metagenomic survey of microbes in honey bee colony collapse disorder. Science 2007, 318, 283–287. [Google Scholar] [CrossRef] [Green Version]
  17. Raymann, K.; Moran, N.A. The role of the gut microbiome in health and disease of adult honey bee workers. Curr. Opin. Insect Sci. 2018, 26, 97–104. [Google Scholar] [CrossRef]
  18. Anderson, K.E.; Ricigliano, V.A. Honey bee gut dysbiosis: A novel context of disease ecology. Vectors Med. Vet. Entomol. Soc. Insects 2017, 22, 125–132. [Google Scholar] [CrossRef]
  19. Evison, S.E.; Jensen, A.B. The biology and prevalence of fungal diseases in managed and wild bees. Ecol. Parasites Parasit. Biol. Control 2018, 26, 105–113. [Google Scholar] [CrossRef] [PubMed]
  20. Forsgren, E. European foulbrood in honey bees. J. Invertebr. Pathol. 2010, 103, S5–S9. [Google Scholar] [CrossRef] [PubMed]
  21. Ebeling, J.; Knispel, H.; Hertlein, G.; Fünfhaus, A.; Genersch, E. Biology of PaeniBacillus larvae, a deadly pathogen of honey bee larvae. Appl. Microbiol. Biotechnol. 2016, 100, 7387–7395. [Google Scholar] [CrossRef] [PubMed]
  22. Fries, I. Nosema ceranae in European honey bees (Apis mellifera). J. Invertebr. Pathol. 2010, 103, S73–S79. [Google Scholar] [CrossRef]
  23. Reybroeck, W.; Daeseleire, E.; De Brabander, H.F.; Herman, L. Antimicrobials in beekeeping. Vet. Microbiol. 2012, 158, 1–11. [Google Scholar] [CrossRef]
  24. Jończyk-Matysiak, E.; Popiela, E.; Owczarek, B.; Hodyra-Stefaniak, K.; Świtała-Jeleń, K.; Łodej, N.; Kula, D.; Neuberg, J.; Migdał, P.; Bagińska, N. Phages in therapy and prophylaxis of american foulbrood–recent implications from practical applications. Front. Microbiol. 2020, 11, 1913. [Google Scholar] [CrossRef] [PubMed]
  25. Mutinelli, F. European legislation governing the authorization of veterinary medicinal products with particular reference to the use of drugs for the control of honey bee diseases. Apiacta 2003, 38, 156–168. [Google Scholar]
  26. Raymann, K.; Shaffer, Z.; Moran, N.A. Antibiotic exposure perturbs the gut microbiota and elevates mortality in honeybees. PLoS Biol. 2017, 15, e2001861. [Google Scholar] [CrossRef]
  27. Lodesani, M.; Costa, C. Limits of chemotherapy in beekeeping: Development of resistance and the problem of residues. Bee World 2005, 86, 102–109. [Google Scholar] [CrossRef]
  28. Brosi, B.J.; Delaplane, K.S.; Boots, M.; de Roode, J.C. Ecological and evolutionary approaches to managing honeybee disease. Nat. Ecol. Evol. 2017, 1, 1250–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Tauber, J.P.; Collins, W.R.; Schwarz, R.S.; Chen, Y.; Grubbs, K.; Huang, Q.; Lopez, D.; Peterson, R.; Evans, J.D. Natural product medicines for honey bees: Perspective and protocols. Insects 2019, 10, 356. [Google Scholar] [CrossRef] [Green Version]
  30. Abou-Shaara, H. Continuous management of Varroa mite in honey bee, Apis mellifera, colonies. Acarina 2014, 22, 149–156. [Google Scholar]
  31. Hamdi, C.; Balloi, A.; Essanaa, J.; Crotti, E.; Gonella, E.; Raddadi, N.; Ricci, I.; Boudabous, A.; Borin, S.; Manino, A.; et al. Gut microbiome dysbiosis and honeybee health. J. Appl. Entomol. 2011, 135, 524–533. [Google Scholar] [CrossRef] [Green Version]
  32. Cariveau, D.P.; Elijah Powell, J.; Koch, H.; Winfree, R.; Moran, N.A. Variation in gut microbial communities and its association with pathogen infection in wild bumble bees (Bombus). ISME J. 2014, 8, 2369–2379. [Google Scholar] [CrossRef]
  33. Daisley, B.A.; Chmiel, J.A.; Pitek, A.P.; Thompson, G.J.; Reid, G. Missing microbes in bees: How systematic depletion of key symbionts erodes immunity. Trends Microbiol. 2020, 28, 1010–1021. [Google Scholar] [CrossRef] [PubMed]
  34. Evans, J.D.; Armstrong, T.-N. Antagonistic interactions between honey bee bacterial symbionts and implications for disease. BMC Ecol. 2006, 6, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Anderson, K.E.; Sheehan, T.H.; Eckholm, B.J.; Mott, B.M.; DeGrandi-Hoffman, G. An emerging paradigm of colony health: Microbial balance of the honey bee and hive (Apis mellifera). Insectes Sociaux 2011, 58, 431. [Google Scholar] [CrossRef]
  36. Wu, M.; Sugimura, Y.; Iwata, K.; Takaya, N.; Takamatsu, D.; Kobayashi, M.; Taylor, D.; Kimura, K.; Yoshiyama, M. Inhibitory effect of gut bacteria from the Japanese honey bee, Apis cerana japonica, against Melissococcus plutonius, the causal agent of European foulbrood disease. J. Insect Sci. Online 2014, 14, 129. [Google Scholar] [CrossRef]
  37. Crotti, E.; Balloi, A.; Hamdi, C.; Sansonno, L.; Marzorati, M.; Gonella, E.; Favia, G.; Cherif, A.; Bandi, C.; Alma, A.; et al. Microbial symbionts: A resource for the management of insect-related problems. Microb. Biotechnol. 2012, 5, 307–317. [Google Scholar] [CrossRef] [Green Version]
  38. Crotti, E.; Sansonno, L.; Prosdocimi, E.M.; Vacchini, V.; Hamdi, C.; Cherif, A.; Gonella, E.; Marzorati, M.; Balloi, A. Microbial symbionts of honeybees: A promising tool to improve honeybee health. New Biotechnol. 2013, 30, 716–722. [Google Scholar] [CrossRef]
  39. Verstraete, W. Microbial ecology and environmental biotechnology. ISME J. 2007, 1, 4–8. [Google Scholar] [CrossRef] [PubMed]
  40. Gaggìa, F.; Baffoni, L.; Alberoni, D. Probiotics for honeybees’ health. In Probiotics and Prebiotics in Animal Health and Food Safety; Di Gioia, D., Biavati, B., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 219–245. ISBN 978-3-319-71950-4. [Google Scholar]
  41. Audisio, M.C. Gram-positive bacteria with probiotic potential for the Apis mellifera L. honey bee: The experience in the Northwest of Argentina. Probiotics Antimicrob. Proteins 2017, 9, 22–31. [Google Scholar] [CrossRef]
  42. Pietropaoli, M.; Carpana, E.; Milito, M.; Palazzetti, M.; Guarducci, M.; Croppi, S.; Formato, G. Use of Lactobacillus plantarum in preventing clinical cases of American and European foulbrood in central Italy. Appl. Sci. 2012, 12–1388. [Google Scholar]
  43. Alberoni, D.; Baffoni, L.; Gaggìa, F.; Ryan, P.; Murphy, K.; Ross, P.; Stanton, C.; Di Gioia, D. Impact of beneficial bacteria supplementation on the gut microbiota, colony development and productivity of Apis mellifera L. Benef. Microbes 2018, 9, 269–278. [Google Scholar] [CrossRef]
  44. Kwong, W.K.; Engel, P.; Koch, H.; Moran, N.A. Genomics and host specialization of honey bee and bumble bee gut symbionts. Proc. Natl. Acad. Sci. USA 2014, 111, 11509–11514. [Google Scholar] [CrossRef] [Green Version]
  45. Kwong, W.K.; Moran, N.A. Gut microbial communities of social bees. Nat. Rev. Microbiol. 2016, 14, 374–384. [Google Scholar] [CrossRef]
  46. Endo, A.; Salminen, S. Honeybees and beehives are rich sources for fructophilic lactic acid bacteria. Syst. Appl. Microbiol. 2013, 36, 444–448. [Google Scholar] [CrossRef]
  47. Engel, P.; Moran, N.A. The gut microbiota of insects—diversity in structure and function. FEMS Microbiol. Rev. 2013, 37, 699–735. [Google Scholar] [CrossRef] [PubMed]
  48. Iorizzo, M.; Pannella, G.; Lombardi, S.J.; Ganassi, S.; Testa, B.; Succi, M.; Sorrentino, E.; Petrarca, S.; De Cristofaro, A.; Coppola, R.; et al. Inter- and intra-species diversity of lactic acid bacteria in Apis mellifera ligustica colonies. Microorganisms 2020, 8, 1578. [Google Scholar] [CrossRef]
  49. Morelli, L.; von Wright, A. Genetics of lactic acid bacteria. In Lactic Acid Bacteria; CRC Press: Boca Raton, FL, USA, 2019; pp. 17–32. ISBN 0-429-05746-6. [Google Scholar]
  50. Lebeer, S.; Vanderleyden, J.; De Keersmaecker, S.C.J. Genes and molecules of lactobacilli supporting probiotic action. Microbiol. Mol. Biol. Rev. MMBR 2008, 72, 728–764. [Google Scholar] [CrossRef] [Green Version]
  51. Klaenhammer, T.R.; Barrangou, R.; Buck, B.L.; Azcarate-Peril, M.A.; Altermann, E. Genomic features of lactic acid bacteria effecting bioprocessing and health. FEMS Microbiol. Rev. 2005, 29, 393–409. [Google Scholar] [CrossRef] [PubMed]
  52. Olofsson, T.C.; Butler, È.; Markowicz, P.; Lindholm, C.; Larsson, L.; Vásquez, A. Lactic acid bacterial symbionts in honeybees—An unknown key to honey’s antimicrobial and therapeutic activities. Int. Wound J. 2016, 13, 668–679. [Google Scholar] [CrossRef]
  53. Endo, A.; Futagawa-Endo, Y.; Dicks, L.M.T. Isolation and characterization of fructophilic lactic acid bacteria from fructose-rich niches. Syst. Appl. Microbiol. 2009, 32, 593–600. [Google Scholar] [CrossRef]
  54. Filannino, P.; Di Cagno, R.; Tlais, A.Z.A.; Cantatore, V.; Gobbetti, M. Fructose-rich niches traced the evolution of lactic acid bacteria toward fructophilic Species. Crit. Rev. Microbiol. 2019, 45, 65–81. [Google Scholar] [CrossRef]
  55. Gaggia, F.; Baffoni, L.; Stenico, V.; Alberoni, D.; Buglione, E.; Lilli, A.; Di Gioia, D.; Porrini, C. Microbial investigation on honey bee larvae showing atypical symptoms of European foulbrood. Bull. Insectol. 2015, 68, 321–327. [Google Scholar]
  56. Olofsson, T.C.; Vásquez, A. Detection and identification of a novel lactic acid bacterial flora within the honey stomach of the honeybee Apis mellifera. Curr. Microbiol. 2008, 57, 356–363. [Google Scholar] [CrossRef] [PubMed]
  57. Cilia, G.; Fratini, F.; Tafi, E.; Turchi, B.; Mancini, S.; Sagona, S.; Nanetti, A.; Cerri, D.; Felicioli, A. Microbial profile of the ventriculum of honey bee (Apis mellifera ligustica Spinola, 1806) fed with veterinary drugs, dietary supplements and non-protein amino acids. Vet. Sci. 2020, 7, 76. [Google Scholar] [CrossRef] [PubMed]
  58. Cilia, G.; Fratini, F.; Tafi, E.; Mancini, S.; Turchi, B.; Sagona, S.; Cerri, D.; Felicioli, A.; Nanetti, A. Changes of western honey bee Apis mellifera ligustica (Spinola, 1806) ventriculus microbial profile related to their in-hive tasks. J. Apic. Res. 2021, 60, 198–202. [Google Scholar] [CrossRef]
  59. Royan, M. Mechanisms of probiotic action in the honeybee. Crit. Rev. Eukaryot. Gene Expr. 2019, 29, 95–103. [Google Scholar] [CrossRef] [PubMed]
  60. Jing, T.-Z.; Qi, F.-H.; Wang, Z.-Y. Most dominant roles of insect gut bacteria: Digestion, detoxification, or essential nutrient provision? Microbiome 2020, 8, 38. [Google Scholar] [CrossRef] [Green Version]
  61. Quinto, E.J.; Jiménez, P.; Caro, I.; Tejero, J.; Mateo, J.; Girbés, T. Probiotic lactic acid bacteria: A review. Food Nutr. 2014, 5, 1765–1775. [Google Scholar] [CrossRef] [Green Version]
  62. Ayivi, R.D.; Gyawali, R.; Krastanov, A.; Aljaloud, S.O.; Worku, M.; Tahergorabi, R.; da Silva, R.C.; Ibrahim, S.A. Lactic acid bacteria: Food safety and human health applications. Dairy 2020, 1, 202–232. [Google Scholar] [CrossRef]
  63. Mora-Villalobos, J.A.; Montero-Zamora, J.; Barboza, N.; Rojas-Garbanzo, C.; Usaga, J.; Redondo-Solano, M.; Schroedter, L.; Olszewska-Widdrat, A.; López-Gómez, J.P. Multi-product lactic acid bacteria fermentations: A review. Fermentation 2020, 6, 23. [Google Scholar] [CrossRef] [Green Version]
  64. Pachla, A.; Ptaszyńska, A.A.; Wicha, M.; Kunat, M.; Wydrych, J.; Oleńska, E.; Małek, W. Insight into probiotic properties of lactic acid bacterial endosymbionts of Apis mellifera L. derived from the polish apiary. Saudi J. Biol. Sci. 2021, 28, 1890–1899. [Google Scholar] [CrossRef]
  65. Audisio, M.; Benítez-Ahrendts, M. Lactobacillus johnsonii CRL1647, isolated from Apis mellifera L. bee-gut, exhibited a beneficial effect on honeybee colonies. Benef. Microbes 2011, 2, 29–34. [Google Scholar] [CrossRef] [PubMed]
  66. Audisio, M.C.; Sabate, D.C.; Benítez-Ahrendts, M.R. Effect of Lactobacillus johnsonii CRL1647 on different parameters of honeybee colonies and bacterial populations of the bee gut. Benef. Microbes 2015, 6, 687–695. [Google Scholar] [CrossRef]
  67. Maggi, M.; Negri, P.; Plischuk, S.; Szawarski, N.; De Piano, F.; De Feudis, L.; Eguaras, M.; Audisio, C. Effects of the organic acids produced by a lactic acid bacterium in Apis mellifera colony development, Nosema ceranae control and fumagillin efficiency. Vet. Microbiol. 2013, 167, 474–483. [Google Scholar] [CrossRef] [PubMed]
  68. Fanciotti, M.N.; Tejerina, M.; Benítez-Ahrendts, M.R.; Audisio, M.C. Honey yield of different commercial apiaries treated with Lactobacillus salivarius A3iob, a new bee-probiotic strain. Benef. Microbes 2018, 9, 291–298. [Google Scholar] [CrossRef]
  69. Máchová, M.; Rada, V.; Huk, J.; Smékal, F. Development of probiotics for bees. Apiacta 1997, 4, 99–111. [Google Scholar]
  70. Pătruică, S.; Mot, D. The Effect of Using prebiotic and probiotic products on intestinal micro-flora of the honeybee (Apis mellifera carpatica). Bull. Entomol. Res. 2012, 102, 619–623. [Google Scholar] [CrossRef]
  71. Pătruică, S.; Dumitrescu, G.; Popescu, R.; Filimon, N.M. The effect of prebiotic and probiotic products used in feed to stimulate the bee colony (Apis mellifera) on intestines of working bees. J. Food Agric. Environ. 2013, 11, 2461–2464. [Google Scholar]
  72. Pătruică, S.; Huțu, I. Economic benefits of using prebiotic and probiotic products as supplements in stimulation feeds administered to bee colonies. Turk. J. Vet. Anim. Sci. 2013, 37, 259–263. [Google Scholar]
  73. Kaznowski, A.; Szymas, B.; Jazdzinska, E.; Kazimierczak, M.; Paetz, H.; Mokracka, J. The effects of probiotic supplementation on the content of intestinal microflora and chemical composition of worker honey bees (Apis mellifera). J. Apic. Res. 2005, 44, 10–14. [Google Scholar] [CrossRef]
  74. Maruščáková, I.C.; Schusterová, P.; Bielik, B.; Toporčák, J.; Bíliková, K.; Mudroňová, D. Effect of application of probiotic pollen suspension on immune response and gut microbiota of honey bees (Apis mellifera). Probiotics Antimicrob. Proteins 2020, 12, 929–936. [Google Scholar] [CrossRef]
  75. Daisley, B.A.; Pitek, A.P.; Chmiel, J.A.; Gibbons, S.; Chernyshova, A.M.; Al, K.F.; Faragalla, K.M.; Burton, J.P.; Thompson, G.J.; Reid, G. Lactobacillus spp. attenuate antibiotic-induced immune and microbiota dysregulation in honey bees. Commun. Biol. 2020, 3, 534. [Google Scholar] [CrossRef]
  76. Evans, J.D.; Lopez, D.L. Bacterial probiotics induce an immune response in the honey bee (Hymenoptera: Apidae). J. Econ. Entomol. 2004, 97, 752–756. [Google Scholar] [CrossRef]
  77. Tlak Gajger, I.; Vlainić, J.; Šoštarić, P.; Prešern, J.; Bubnič, J.; Smodiš Škerl, M.I. Effects on some therapeutical, biochemical, and immunological parameters of honey bee (Apis mellifera) exposed to probiotic treatments, in field and laboratory conditions. Insects 2020, 11, 638. [Google Scholar] [CrossRef]
  78. Janashia, I.; Alaux, C. Specific immune stimulation by endogenous bacteria in honey bees (Hymenoptera: Apidae). J. Econ. Entomol. 2016, 109, 1474–1477. [Google Scholar] [CrossRef]
  79. Rokop, Z.P.; Horton, M.A.; Newton, I.L.G. Interactions between cooccurring lactic acid bacteria in honey bee hives. Appl. Environ. Microbiol. 2015, 81, 7261–7270. [Google Scholar] [CrossRef] [Green Version]
  80. Yoshiyama, M.; Wu, M.; Sugimura, Y.; Takaya, N.; Kimoto-Nira, H.; Suzuki, C. Inhibition of PaeniBacillus Larvae by lactic acid bacteria isolated from fermented materials. J. Invertebr. Pathol. 2013, 112, 62–67. [Google Scholar] [CrossRef]
  81. Feldhaar, H.; Otti, O. Pollutants and their interaction with diseases of social Hymenoptera. Insects 2020, 11, 153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zwolak, A.; Sarzyńska, M.; Szpyrka, E.; Stawarczyk, K. Sources of soil pollution by heavy metals and their accumulation in vegetables: A review. Water Air Soil Pollut. 2019, 230, 164. [Google Scholar] [CrossRef] [Green Version]
  83. Singh, J.; Kalamdhad, A.S. Effects of heavy metals on soil, plants, human health and aquatic life. Int. J. Res. Chem. Environ. 2011, 1, 15–21. [Google Scholar]
  84. Briffa, J.; Sinagra, E.; Blundell, R. Heavy metal pollution in the environment and their toxicological effects on humans. Heliyon 2020, 6, e04691. [Google Scholar] [CrossRef]
  85. Rothman, J.A.; Leger, L.; Kirkwood, J.S.; McFrederick, Q.S. Cadmium and selenate exposure affects the honey bee microbiome and metabolome, and bee-associated bacteria show potential for bioaccumulation. Appl. Environ. Microbiol. 2019, 85, e01411-19. [Google Scholar] [CrossRef] [Green Version]
  86. Hladun, K.R.; Di, N.; Liu, T.-X.; Trumble, J.T. Metal contaminant accumulation in the hive: Consequences for whole-colony health and brood production in the honey bee (Apis mellifera L.). Environ. Toxicol. Chem. 2016, 35, 322–329. [Google Scholar] [CrossRef]
  87. Gauthier, M.; Aras, P.; Jumarie, C.; Boily, M. Low dietary levels of Al, Pb and Cd may affect the non-enzymatic antioxidant capacity in caged honey bees (Apis mellifera). Chemosphere 2016, 144, 848–854. [Google Scholar] [CrossRef]
  88. Polykretis, P.; Delfino, G.; Petrocelli, I.; Cervo, R.; Tanteri, G.; Montori, G.; Perito, B.; Branca, J.J.V.; Morucci, G.; Gulisano, M. Evidence of immunocompetence reduction induced by cadmium exposure in honey bees (Apis mellifera). Environ. Pollut. 2016, 218, 826–834. [Google Scholar] [CrossRef]
  89. Dabour, K.; Al Naggar, Y.; Masry, S.; Naiem, E.; Giesy, J.P. Cellular alterations in midgut cells of honey bee workers (Apis Mellifera L.) exposed to sublethal concentrations of CdO or PbO nanoparticles or their binary mixture. Sci. Total Environ. 2019, 651, 1356–1367. [Google Scholar] [CrossRef]
  90. Wang, K. Tolerance of cultivated plants to cadmium and their utilization in polluted farmland soils. Acta Biotechnol. 2002, 22, 189–198. [Google Scholar] [CrossRef]
  91. Di, N.; Hladun, K.R.; Zhang, K.; Liu, T.-X.; Trumble, J.T. Laboratory bioassays on the impact of cadmium, copper and lead on the development and survival of honeybee (Apis mellifera L.) larvae and foragers. Chemosphere 2016, 152, 530–538. [Google Scholar] [CrossRef] [Green Version]
  92. Conti, M.E.; Botrè, F. Honeybees and their products as potential bioindicators of heavy metals contamination. Environ. Monit. Assess. 2001, 69, 267–282. [Google Scholar] [CrossRef]
  93. Zarić, N.M.; Ilijević, K.; Stanisavljević, L.; Gržetić, I. Metal concentrations around thermal power plants, rural and urban areas using honeybees (Apis mellifera L.) as bioindicators. Int. J. Environ. Sci. Technol. 2016, 13, 413–422. [Google Scholar] [CrossRef] [Green Version]
  94. Di, N.; Zhang, K.; Hladun, K.R.; Rust, M.; Chen, Y.-F.; Zhu, Z.-Y.; Liu, T.-X.; Trumble, J.T. Joint effects of cadmium and copper on Apis mellifera forgers and larvae. Comp. Biochem. Physiol. Part C 2020, 237, 108839. [Google Scholar] [CrossRef] [PubMed]
  95. Hladun, K.R.; Smith, B.H.; Mustard, J.A.; Morton, R.R.; Trumble, J.T. Selenium toxicity to honey bee (Apis mellifera L.) pollinators: Effects on behaviors and survival. PLoS ONE 2012, 7, e34137. [Google Scholar] [CrossRef] [PubMed]
  96. Hladun, K.R.; Kaftanoglu, O.; Parker, D.R.; Tran, K.D.; Trumble, J.T. Effects of selenium on development, survival, and accumulation in the honeybee (Apis mellifera L.). Environ. Toxicol. Chem. 2013, 32, 2584–2592. [Google Scholar] [CrossRef] [PubMed]
  97. Søvik, E.; Perry, C.J.; LaMora, A.; Barron, A.B.; Ben-Shahar, Y. Negative impact of manganese on honeybee foraging. Biol. Lett. 2015, 11, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Burden, C.M.; Morgan, M.O.; Hladun, K.R.; Amdam, G.V.; Trumble, J.J.; Smith, B.H. Acute sublethal exposure to toxic heavy metals alters honey bee (Apis mellifera) feeding behavior. Sci. Rep. 2019, 9, 4253. [Google Scholar] [CrossRef]
  99. Gizaw, G.; Kim, Y.; Moon, K.; Choi, J.B.; Kim, Y.H.; Park, J.K. Effect of environmental heavy metals on the expression of detoxification-related genes in honey bee Apis mellifera. Apidologie 2020, 51, 664–674. [Google Scholar] [CrossRef]
  100. AL Naggar, Y.; Dabour, K.; Masry, S.; Sadek, A.; Naiem, E.; Giesy, J.P. Sublethal effects of chronic exposure to CdO or PbO nanoparticles or their binary mixture on the honey bee (Apis mellifera L.). Environ. Sci. Pollut. Res. 2020, 27, 19004–19015. [Google Scholar] [CrossRef]
  101. Jumarie, C.; Aras, P.; Boily, M. Mixtures of herbicides and metals affect the redox system of honey bees. Chemosphere 2017, 168, 163–170. [Google Scholar] [CrossRef] [PubMed]
  102. Kinoshita, H. Biosorption of heavy metals by lactic acid bacteria for detoxification. In Lactic Acid Bacteria: Methods and Protocols; Kanauchi, M., Ed.; Springer: New York, NY, USA, 2019; pp. 145–157. ISBN 978-1-4939-8907-2. [Google Scholar]
  103. Bhakta, J.N.; Ohnishi, K.; Munekage, Y.; Iwasaki, K.; Wei, M.Q. Characterization of lactic acid bacteria-based probiotics as potential heavy metal sorbents. J. Appl. Microbiol. 2012, 112, 1193–1206. [Google Scholar] [CrossRef] [PubMed]
  104. Mohammadi, M.; Shadnoush, M.; Sohrabvandi, S.; Yousefi, M.; Khorshidian, N.; Mortazavian, A.M. Probiotics as potential detoxification tools for mitigation of pesticides: A mini review. Int. J. Food Sci. Technol. 2021, 56, 2078–2087. [Google Scholar] [CrossRef]
  105. Zhai, Q.; Yin, R.; Yu, L.; Wang, G.; Tian, F.; Yu, R.; Zhao, J.; Liu, X.; Chen, Y.Q.; Zhang, H. Screening of lactic acid bacteria with potential protective effects against cadmium toxicity. Food Control 2015, 54, 23–30. [Google Scholar] [CrossRef]
  106. Lin, D.; Ji, R.; Wang, D.; Xiao, M.; Zhao, J.; Zou, J.; Li, Y.; Qin, T.; Xing, B.; Chen, Y.; et al. The research progress in mechanism and influence of biosorption between lactic acid bacteria and Pb(II): A review. Crit. Rev. Food Sci. Nutr. 2019, 59, 395–410. [Google Scholar] [CrossRef] [PubMed]
  107. Ibrahim, F.; Halttunen, T.; Tahvonen, R.; Salminen, S. Probiotic bacteria as potential detoxification tools: Assessing their heavy metal binding isotherms. Can. J. Microbiol. 2006, 52, 877–885. [Google Scholar] [CrossRef] [PubMed]
  108. Nowak, A.; Szczuka, D.; Górczyńska, A.; Motyl, I.; Kręgiel, D. Characterization of Apis mellifera gastrointestinal microbiota and lactic acid bacteria for honeybee protection-A review. Cells 2021, 10, 701. [Google Scholar] [CrossRef] [PubMed]
  109. Abdel-Megeed, R.M. Probiotics: A promising generation of heavy metal detoxification. Biol. Trace Elem. Res. 2021, 199, 2406–2413. [Google Scholar] [CrossRef]
  110. Duan, H.; Yu, L.; Tian, F.; Zhai, Q.; Fan, L.; Chen, W. Gut microbiota: A target for heavy metal toxicity and a probiotic protective strategy. Sci. Total Environ. 2020, 742, 140429. [Google Scholar] [CrossRef]
  111. Tian, F.; Xiao, Y.; Li, X.; Zhai, Q.; Wang, G.; Zhang, Q.; Zhang, H.; Chen, W. Protective effects of Lactobacillus plantarum CCFM8246 against copper toxicity in mice. PLoS ONE 2015, 10, e0143318. [Google Scholar] [CrossRef]
  112. Kumar, N.; Kumar, V.; Panwar, R.; Ram, C. Efficacy of Indigenous probiotic Lactobacillus strains to reduce cadmium bioaccessibility—An in vitro digestion model. Environ. Sci. Pollut. Res. 2017, 24, 1241–1250. [Google Scholar] [CrossRef]
  113. George, F.; Mahieux, S.; Daniel, C.; Titécat, M.; Beauval, N.; Houcke, I.; Neut, C.; Allorge, D.; Borges, F.; Jan, G.; et al. Assessment of Pb(II), Cd(II), and Al(III) removal capacity of bacteria from food and gut ecological niches: Insights into biodiversity to limit intestinal biodisponibility of toxic metals. Microorganisms 2021, 9, 456. [Google Scholar] [CrossRef]
  114. Coryell, M.; McAlpine, M.; Pinkham, N.V.; McDermott, T.R.; Walk, S.T. The gut microbiome is required for full protection against acute arsenic toxicity in mouse models. Nat. Commun. 2018, 9, 5424. [Google Scholar] [CrossRef] [PubMed]
  115. Zhai, Q.; Wang, G.; Zhao, J.; Liu, X.; Tian, F.; Zhang, H.; Chen, W. Protective effects of Lactobacillus Plantarum CCFM8610 against acute cadmium toxicity in mice. Appl. Environ. Microbiol. 2013, 79, 1508–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zhai, Q.; Narbad, A.; Chen, W. Dietary strategies for the treatment of cadmium and lead toxicity. Nutrients 2015, 7, 552–571. [Google Scholar] [CrossRef] [Green Version]
  117. Zhai, Q.; Yu, L.; Li, T.; Zhu, J.; Zhang, C.; Zhao, J.; Zhang, H.; Chen, W. Effect of dietary probiotic supplementation on intestinal microbiota and physiological conditions of nile tilapia (Oreochromis niloticus) under waterborne cadmium exposure. Antonie Van Leeuwenhoek 2017, 110, 501–513. [Google Scholar] [CrossRef] [PubMed]
  118. Kumar, N.; Kumari, V.; Ram, C.; Thakur, K.; Tomar, S.K. Bio-prospectus of cadmium bioadsorption by lactic acid bacteria to mitigate health and environmental impacts. Appl. Microbiol. Biotechnol. 2018, 102, 1599–1615. [Google Scholar] [CrossRef] [PubMed]
  119. Fisher II, A.; DeGrandi-Hoffman, G.; Smith, B.H.; Johnson, M.; Kaftanoglu, O.; Cogley, T.; Fewell, J.H.; Harrison, J.F. Colony field test reveals dramatically higher toxicity of a widely-used mito-toxic fungicide on honey bees (Apis mellifera). Environ. Pollut. 2021, 269, 115964. [Google Scholar] [CrossRef] [PubMed]
  120. Pettis, J.S.; vanEngelsdorp, D.; Johnson, J.; Dively, G. Pesticide exposure in honey bees results in increased levels of the gut pathogen Nosema. Naturwissenschaften 2012, 99, 153–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Straub, L.; Williams, G.R.; Vidondo, B.; Khongphinitbunjong, K.; Retschnig, G.; Schneeberger, A.; Chantawannakul, P.; Dietemann, V.; Neumann, P. Neonicotinoids and ectoparasitic mites synergistically impact honeybees. Sci. Rep. 2019, 9, 8159. [Google Scholar] [CrossRef] [PubMed]
  122. Tesovnik, T.; Zorc, M.; Ristanić, M.; Glavinić, U.; Stevanović, J.; Narat, M.; Stanimirović, Z. Exposure of honey bee larvae to thiamethoxam and its interaction with Nosema Ceranae infection in adult honey bees. Environ. Pollut. 2020, 256, 113443. [Google Scholar] [CrossRef] [PubMed]
  123. Glavinic, U.; Tesovnik, T.; Stevanovic, J.; Zorc, M.; Cizelj, I.; Stanimirovic, Z.; Narat, M. Response of adult honey bees treated in larval stage with prochloraz to infection with Nosema ceranae. PeerJ 2019, 7, e6325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Motta, E.V.; Raymann, K.; Moran, N.A. Glyphosate perturbs the gut microbiota of honey bees. Proc. Natl. Acad. Sci. USA 2018, 115, 10305–10310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Motta, E.V.; Moran, N.A. Impact of glyphosate on the honey bee gut microbiota: Effects of intensity, duration, and timing of exposure. Msystems 2020, 5, e00268-20. [Google Scholar] [CrossRef] [PubMed]
  126. Ye, M.-H.; Fan, S.-H.; Li, X.-Y.; Tarequl, I.M.; Yan, C.-X.; Wei, W.-H.; Yang, S.-M.; Zhou, B. Microbiota dysbiosis in honeybee (Apis mellifera L.) larvae infected with brood diseases and foraging bees exposed to agrochemicals. R. Soc. Open Sci. 2021, 8, 201805. [Google Scholar] [PubMed]
  127. Alaux, C.; Brunet, J.-L.; Dussaubat, C.; Mondet, F.; Tchamitchan, S.; Cousin, M.; Brillard, J.; Baldy, A.; Belzunces, L.P.; Le Conte, Y. Interactions between Nosema microspores and a neonicotinoid weaken honeybees (Apis mellifera). Environ. Microbiol. 2010, 12, 774–782. [Google Scholar] [CrossRef] [Green Version]
  128. Vidau, C.; Diogon, M.; Aufauvre, J.; Fontbonne, R.; Viguès, B.; Brunet, J.-L.; Texier, C.; Biron, D.G.; Blot, N.; El Alaoui, H.; et al. Exposure to sublethal doses of fipronil and thiacloprid highly increases mortality of honeybees previously infected by Nosema ceranae. PLoS ONE 2011, 6, e21550. [Google Scholar] [CrossRef] [Green Version]
  129. Claudianos, C.; Ranson, H.; Johnson, R.M.; Biswas, S.; Schuler, M.A.; Berenbaum, M.R.; Feyereisen, R.; Oakeshott, J.G. A deficit of detoxification enzymes: Pesticide sensitivity and environmental response in the honeybee. Insect Mol. Biol. 2006, 15, 615–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Wu, Y.; Zheng, Y.; Chen, Y.; Wang, S.; Chen, Y.; Hu, F.; Zheng, H. Honey bee (Apis mellifera) gut microbiota promotes host endogenous detoxification capability via regulation of P450 gene expression in the digestive tract. Microb. Biotechnol. 2020, 13, 1201–1212. [Google Scholar] [CrossRef] [PubMed]
  131. Peghaire, E.; Moné, A.; Delbac, F.; Debroas, D.; Chaucheyras-Durand, F.; El Alaoui, H. A pediococcus strain to rescue honeybees by decreasing Nosema ceranae- and pesticide-induced adverse effects. Pestic. Biochem. Physiol. 2020, 163, 138–146. [Google Scholar] [CrossRef] [PubMed]
  132. Islam, S.M.A.; Math, R.K.; Cho, K.M.; Lim, W.J.; Hong, S.Y.; Kim, J.M.; Yun, M.G.; Cho, J.J.; Yun, H.D. Organophosphorus hydrolase (OpdB) of Lactobacillus brevis WCP902 from kimchi is able to degrade organophosphorus pesticides. J. Agric. Food Chem. 2010, 58, 5380–5386. [Google Scholar]
  133. Zhang, Y.-H.; Xu, D.; Liu, J.-Q.; Zhao, X.-H. Enhanced degradation of five organophosphorus pesticides in skimmed milk by lactic acid bacteria and its potential relationship with phosphatase production. Food Chem. 2014, 164, 173–178. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, Y.-H.; Xu, D.; Zhao, X.-H.; Song, Y.; Liu, Y.-L.; Li, H.-N. Biodegradation of two organophosphorus pesticides in whole corn silage as affected by the cultured Lactobacillus plantarum. 3 Biotech 2016, 6, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Li, C.; Ma, Y.; Mi, Z.; Huo, R.; Zhou, T.; Hai, H.; Kwok, L.-Y.; Sun, Z.; Chen, Y.; Zhang, H. Screening for Lactobacillus plantarum strains that possess organophosphorus pesticide-degrading activity and metabolomic analysis of phorate degradation. Front. Microbiol. 2018, 9, 2048. [Google Scholar] [CrossRef] [Green Version]
  136. Trinder, M.; Bisanz, J.; Burton, J.; Reid, G. Probiotic lactobacilli: A potential prophylactic treatment for reducing pesticide absorption in humans and wildlife. Benef. Microbes 2015, 6, 841–847. [Google Scholar] [CrossRef] [PubMed]
  137. Trinder, M.; McDowell, T.W.; Daisley, B.A.; Ali, S.N.; Leong, H.S.; Sumarah, M.W.; Reid, G. Probiotic Lactobacillus rhamnosus reduces organophosphate pesticide absorption and toxicity to Drosophila melanogaster. Appl. Environ. Microbiol. 2016, 82, 6204–6213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Bouhafs, L.; Moudilou, E.N.; Exbrayat, J.M.; Lahouel, M.; Idoui, T. Protective effects of probiotic Lactobacillus plantarum BJ0021 on liver and kidney oxidative stress and apoptosis induced by endosulfan in pregnant rats. Ren. Fail. 2015, 37, 1370–1378. [Google Scholar] [CrossRef] [PubMed]
  139. Bagherpour Shamloo, H.; Golkari, S.; Faghfoori, Z.; Movassaghpour, A.; Lotfi, H.; Barzegari, A.; Yari Khosroushahi, A. Lactobacillus casei decreases organophosphorus pesticide diazinon cytotoxicity in human HUVEC cell line. Adv. Pharm. Bull. 2016, 6, 201–210. [Google Scholar] [CrossRef] [PubMed]
  140. Daisley, B.A.; Trinder, M.; McDowell, T.W.; Welle, H.; Dube, J.S.; Ali, S.N.; Leong, H.S.; Sumarah, M.W.; Reid, G. Neonicotinoid-induced pathogen susceptibility is mitigated by Lactobacillus plantarum immune stimulation in a Drosophila melanogaster model. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef] [PubMed]
  141. Pinto, G.; Castro, I.; Miguel, M.; Koblitz, M. Lactic acid bacteria-promising technology for organophosphate degradation in food: A pilot study. LWT 2019, 110, 353–359. [Google Scholar] [CrossRef]
  142. Yuan, S.; Li, C.; Yu, H.; Xie, Y.; Guo, Y.; Yao, W. Screening of lactic acid bacteria for degrading organophosphorus pesticides and their potential protective effects against pesticide toxicity. LWT 2021, 147, 111672. [Google Scholar] [CrossRef]
  143. Chmiel, J.A.; Daisley, B.A.; Pitek, A.P.; Thompson, G.J.; Reid, G. Understanding the effects of sublethal pesticide exposure on honey bees: A role for probiotics as mediators of environmental stress. Front. Ecol. Evol. 2020, 8, 22. [Google Scholar] [CrossRef] [Green Version]
  144. Liu, Q.; Yu, Z.; Tian, F.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Surface components and metabolites of probiotics for regulation of intestinal epithelial barrier. Microb. Cell Factories 2020, 19, 23. [Google Scholar] [CrossRef] [PubMed]
  145. Ren, C.; Zhang, Q.; de Haan, B.J.; Faas, M.M.; Zhang, H.; de Vos, P. Protective effects of lactic acid bacteria on gut epithelial barrier dysfunction are Toll like receptor 2 and protein kinase C dependent. Food Funct. 2020, 11, 1230–1234. [Google Scholar] [CrossRef] [PubMed]
  146. Limoli, D.H.; Jones, C.J.; Wozniak, D.J. Bacterial extracellular polysaccharides in biofilm formation and function. Microbiol. Spectr. 2015, 3. [Google Scholar] [CrossRef] [Green Version]
  147. Lembre, P.; Lorentz, C.; Di Martino, P. Exopolysaccharides of the biofilm matrix: A complex biophysical world. Complex World Polysacch. 2012, 371–392. [Google Scholar]
  148. Douglas, A.E. Multiorganismal insects: Diversity and function of resident microorganisms. Annu. Rev. Entomol. 2015, 60, 17–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zheng, H.; Perreau, J.; Powell, J.E.; Han, B.; Zhang, Z.; Kwong, W.K.; Tringe, S.G.; Moran, N.A. Division of labor in honey bee gut microbiota for plant polysaccharide digestion. Proc. Natl. Acad. Sci. USA 2019, 116, 25909–25916. [Google Scholar] [CrossRef] [PubMed]
  150. Stanley, D.; Rejzek, M.; Naested, H.; Smedley, M.; Otero, S.; Fahy, B.; Thorpe, F.; Nash, R.J.; Harwood, W.; Svensson, B.; et al. The role of alpha-glucosidase in germinating barley grains. Plant Physiol. 2011, 155, 932–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Haydak, M.H. Honey bee nutrition. Annu. Rev. Entomol. 1970, 15, 143–156. [Google Scholar] [CrossRef]
  152. Johnson, R.M. Honey bee toxicology. Annu. Rev. Entomol. 2015, 60, 415–434. [Google Scholar] [CrossRef] [Green Version]
  153. Zheng, H.; Nishida, A.; Kwong, W.K.; Koch, H.; Engel, P.; Steele, M.I.; Moran, N.A. Metabolism of toxic sugars by strains of the bee gut symbiont Gilliamella apicola. mBio 2016, 7, e01326-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Iorizzo, M.; Lombardi, S.J.; Ganassi, S.; Testa, B.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; Vergalito, F.; Cozzolino, A.; et al. Antagonistic activity against Ascosphaera apis and functional properties of Lactobacillus kunkeei strains. Antibiotics 2020, 9, 262. [Google Scholar] [CrossRef] [PubMed]
  155. Iorizzo, M.; Testa, B.; Lombardi, S.J.; Ganassi, S.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; Vergalito, F.; Cozzolino, A.; et al. Antimicrobial activity against PaeniBacillus larvae and functional properties of Lactiplantibacillus plantarum Strains: Potential benefits for honeybee health. Antibiotics 2020, 9, 442. [Google Scholar] [CrossRef]
  156. Lee, F.J.; Rusch, D.B.; Stewart, F.J.; Mattila, H.R.; Newton, I.L.G. Saccharide breakdown and fermentation by the honey bee gut microbiome. Environ. Microbiol. 2015, 17, 796–815. [Google Scholar] [CrossRef] [PubMed]
  157. Simone-Finstrom, M.; Li-Byarlay, H.; Huang, M.H.; Strand, M.K.; Rueppell, O.; Tarpy, D.R. Migratory management and environmental conditions affect lifespan and oxidative stress in honey bees. Sci. Rep. 2016, 6, 32023. [Google Scholar] [CrossRef] [PubMed]
  158. Li, Z.; Hou, M.; Qiu, Y.; Zhao, B.; Nie, H.; Su, S. Changes in antioxidant enzymes activity and metabolomic profiles in the guts of honey bee (Apis mellifera) larvae infected with Ascosphaera apis. Insects 2020, 11, 419. [Google Scholar] [CrossRef] [PubMed]
  159. OLGUN, T.; DAYIOĞLU, M.; TAŞKIRAN, N.Ö. Pesticide and pathogen induced oxidative stress in honey bees (Apis mellifera L.). Mellifera 2020, 20, 32–52. [Google Scholar]
  160. Paris, L.; Roussel, M.; Pereira, B.; Delbac, F.; Diogon, M. Disruption of oxidative balance in the gut of the western honeybee Apis mellifera exposed to the intracellular parasite Nosema ceranae and to the insecticide fipronil. Microb. Biotechnol. 2017, 10, 1702–1717. [Google Scholar] [CrossRef] [PubMed]
  161. Weirich, G.F.; Collins, A.M.; Williams, V.P. Antioxidant enzymes in the honey bee, Apis mellifera. Apidologie 2002, 33, 3–14. [Google Scholar] [CrossRef] [Green Version]
  162. Nikolić, T.V.; Purać, J.; Orčić, S.; Kojić, D.; Vujanović, D.; Stanimirović, Z.; Gržetić, I.; Ilijević, K.; Šikoparija, B.; Blagojević, D.P. Environmental effects on superoxide dismutase and catalase activity and expression in honey bee. Arch. Insect Biochem. Physiol. 2015, 90, 181–194. [Google Scholar]
  163. Collins, A.M.; Williams, V.; Evans, J.D. Sperm storage and antioxidative enzyme expression in the honey bee, Apis mellifera. Insect Mol. Biol. 2004, 13, 141–146. [Google Scholar] [CrossRef]
  164. Balieira, K.V.B.; Mazzo, M.; Bizerra, P.F.V.; de Guimarães, A.R.J.S.; Nicodemo, D.; Mingatto, F.E. Imidacloprid-induced oxidative stress in honey bees and the antioxidant action of caffeine. Apidologie 2018, 49, 562–572. [Google Scholar] [CrossRef] [Green Version]
  165. Belsky, J.; Joshi, N.K. Impact of biotic and abiotic stressors on managed and feral bees. Insects 2019, 10, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Drossart, M.; Gérard, M. Beyond the decline of wild bees: Optimizing conservation measures and bringing together the actors. Insects 2020, 11, 649. [Google Scholar] [CrossRef]
  167. Li, X.; Ma, W.; Shen, J.; Long, D.; Feng, Y.; Su, W.; Xu, K.; Du, Y.; Jiang, Y. Tolerance and response of two honeybee species Apis cerana and Apis mellifera to high temperature and relative humidity. PLoS ONE 2019, 14, e0217921. [Google Scholar] [CrossRef] [Green Version]
  168. Mucci, C.A.; Ramirez, L.; Giffoni, R.S.; Lamattina, L. Cold stress induces specific antioxidant responses in honey bee brood. Apidologie 2021, 52, 596–607. [Google Scholar] [CrossRef]
  169. Dussaubat, C.; Brunet, J.-L.; Higes, M.; Colbourne, J.K.; Lopez, J.; Choi, J.-H.; Martín-Hernández, R.; Botías, C.; Cousin, M.; McDonnell, C.; et al. Gut pathology and responses to the microsporidium Nosema ceranae in the honey bee Apis mellifera. PLoS ONE 2012, 7, e37017. [Google Scholar] [CrossRef] [Green Version]
  170. Feng, T.; Wang, J. Oxidative stress tolerance and antioxidant capacity of lactic acid bacteria as probiotic: A systematic review. Gut Microbes 2020, 12, 1801944. [Google Scholar] [CrossRef]
  171. Mishra, V.; Shah, C.; Mokashe, N.; Chavan, R.; Yadav, H.; Prajapati, J. Probiotics as potential antioxidants: A systematic review. J. Agric. Food Chem. 2015, 63, 3615–3626. [Google Scholar] [CrossRef]
  172. Kullisaar, T.; Zilmer, M.; Mikelsaar, M.; Vihalemm, T.; Annuk, H.; Kairane, C.; Kilk, A. Two antioxidative lactobacilli strains as promising probiotics. Int. J. Food Microbiol. 2002, 72, 215–224. [Google Scholar] [CrossRef]
  173. Amaretti, A.; Di Nunzio, M.; Pompei, A.; Raimondi, S.; Rossi, M.; Bordoni, A. Antioxidant properties of potentially probiotic bacteria: In vitro and in vivo activities. Appl. Microbiol. Biotechnol. 2013, 97, 809–817. [Google Scholar] [CrossRef]
  174. Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant properties of probiotic bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef] [PubMed]
  175. Nowak, A.; Paliwoda, A.; Błasiak, J. Anti-proliferative, pro-apoptotic and anti-oxidative activity of Lactobacillus and Bifidobacterium strains: A review of mechanisms and therapeutic perspectives. Crit. Rev. Food Sci. Nutr. 2019, 59, 3456–3467. [Google Scholar] [CrossRef] [PubMed]
  176. Tejerina, M.R.; Cabana, M.J.; Carrillo, L.; Benitez-Ahrendts, M.R. Effect of lactic bacteria on Ascosphaera apisand and A. atra. Asian J. Agric. Food Sci. 2018, 6. [Google Scholar] [CrossRef] [Green Version]
  177. Bielik, B.; Molnár, L.; Vrabec, V.; Andrášiová, R.; Maruščáková, I.C.; Nemcová, R.; Toporčák, J.; Mudroňová, D. Biofilm-forming lactic acid bacteria of honey bee origin intended for potential probiotic use. Acta Vet. Hung. 2021, 68, 345–353. [Google Scholar] [CrossRef] [PubMed]
  178. Iorizzo, M.; Testa, B.; Ganassi, S.; Lombardi, S.J.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; Vergalito, F.; Cozzolino, A.; et al. Probiotic properties and potentiality of Lactiplantibacillus plantarum strains for the biological control of chalkbrood disease. J. Fungi 2021, 7, 379. [Google Scholar] [CrossRef]
  179. Tejerina, M.R.; Cabana, M.J.; Benitez-Ahrendts, M.R. Strains of Lactobacillus Spp. reduce chalkbrood in Apis mellifera. J. Invertebr. Pathol. 2021, 178, 107521. [Google Scholar] [CrossRef] [PubMed]
  180. Mudroňová, D.; Toporčák, J.; Nemcová, R.; Gancarčíková, S.; Hajdučková, V.; Rumanovská, K. Lactobacillus Sp. as a potential probiotic for the prevention of PaeniBacillus larvae infection in honey bees. J. Apic. Res. 2011, 50, 323–324. [Google Scholar] [CrossRef]
  181. Kačániová, M.; Gasper, J.; Terentjeva, M.; Kunová, S.; Kluz, M.; Puchalski, C. Antibacterial activity of bees gut lactobacilli against PaeniBacillus larvae in vitro. Adv. Res. Life Sci. 2018, 2, 7–10. [Google Scholar] [CrossRef] [Green Version]
  182. Killer, J.; Dubná, S.; Sedláček, I.; Švec, P. Lactobacillus Apis sp. nov., from the stomach of honeybees (Apis mellifera), having an in vitro inhibitory effect on the causative agents of American and European foulbrood. Int. J. Syst. Evol. Microbiol. 2014, 64, 152–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Forsgren, E.; Olofsson, T.C.; Váasquez, A.; Fries, I. Novel lactic acid bacteria inhibiting PaeniBacillus Larvae in honey bee larvae. Apidologie 2010, 41, 99–108. [Google Scholar] [CrossRef] [Green Version]
  184. Al-Ghamdi, A.; Ali Khan, K.; Javed Ansari, M.; Almasaudi, S.B.; Al-Kahtani, S. Effect of gut bacterial isolates from Apis mellifera jemenitica on PaeniBacillus larvae infected bee larvae. Saudi J. Biol. Sci. 2018, 25, 383–387. [Google Scholar] [CrossRef]
  185. Lamei, S.; Stephan, J.G.; Riesbeck, K.; Vasquez, A.; Olofsson, T.; Nilson, B.; de Miranda, J.R.; Forsgren, E. The secretome of honey bee-specific lactic acid bacteria inhibits PaeniBacillus larvae growth. J. Apic. Res. 2019, 58, 405–412. [Google Scholar] [CrossRef] [Green Version]
  186. Daisley, B.A.; Pitek, A.P.; Chmiel, J.A.; Al, K.F.; Chernyshova, A.M.; Faragalla, K.M.; Burton, J.P.; Thompson, G.J.; Reid, G. Novel probiotic approach to counter PaeniBacillus larvae infection in honey bees. ISME J. 2020, 14, 476–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Arredondo, D.; Castelli, L.; Porrini, M.P.; Garrido, P.M.; Eguaras, M.J.; Zunino, P.; Antunez, K. Lactobacillus kunkeei strains decreased the infection by honey bee pathogens PaeniBacillus larvae and Nosema ceranae. Benef. Microbes 2018, 9, 279–290. [Google Scholar] [CrossRef] [PubMed]
  188. Dimov, S.G.; Guyrova, A.; Vladimirova, A.; Dimitrov, M.; Peykov, S.; Strateva, T. WGS-based characterization of the potentially beneficial enterococcus faecium efd from a beehive. Mol. Biol. Rep. 2020, 47, 6445–6449. [Google Scholar] [CrossRef] [PubMed]
  189. Gyurova, A.; Vladimirova, A.; Peykov, S.; Dimitrov, M.; Strateva, T.; Dimov, S.G. Characterization of Enterococcus durans EDD2, a strain from beehives with inhibitory activity against PaeniBacillus larvae. J. Apic. Res. 2021, 1–14. [Google Scholar] [CrossRef]
  190. Peykov, S.; Vladimirova, A.; Guyrova, A.; Dimitrov, M.; Strateva, T.; Dimov, S.G. Draft genome sequences of Enterococcus durans EDD2 strain associated with honeybees. AIMS Agric. Food 2020, 5, 288–291. [Google Scholar] [CrossRef]
  191. Jaouani, I.; Abbassi, M.S.; Alessandria, V.; Bouraoui, J.; Ben Salem, R.; Kilani, H.; Mansouri, R.; Messadi, L.; Cocolin, L. High inhibition of PaeniBacillus larvae and Listeria monocytogenes by Enterococcus isolated from different sources in Tunisia and identification of their bacteriocin genes. Lett. Appl. Microbiol. 2014, 59, 17–25. [Google Scholar] [CrossRef] [PubMed]
  192. Vásquez, A.; Forsgren, E.; Fries, I.; Paxton, R.J.; Flaberg, E.; Szekely, L.; Olofsson, T.C. Symbionts as major modulators of insect health: Lactic acid bacteria and honeybees. PLoS ONE 2012, 7, e33188. [Google Scholar] [CrossRef]
  193. Zendo, T.; Ohashi, C.; Maeno, S.; Piao, X.; Salminen, S.; Sonomoto, K.; Endo, A. Kunkecin A, a new nisin variant bacteriocin produced by the fructophilic lactic acid bacterium, ApiLactobacillus Kunkeei FF30-6 isolated from honey bees. Front. Microbiol. 2020, 11, 571903. [Google Scholar] [CrossRef] [PubMed]
  194. Tejerina, M.R.; Benítez-Ahrendts, M.R.; Audisio, M.C. Lactobacillus salivarius A3iob reduces the incidence of Varroa destructor and Nosema spp. in commercial apiaries located in the northwest of Argentina. Probiotics Antimicrob. Proteins 2020, 12, 1360–1369. [Google Scholar] [CrossRef] [PubMed]
  195. Baffoni, L.; Gaggìa, F.; Alberoni, D.; Cabbri, R.; Nanetti, A.; Biavati, B.; Di Gioia, D. Effect of dietary supplementation of Bifidobacterium and Lactobacillus strains in Apis mellifera L. against Nosema ceranae. Benef. Microbes 2016, 7, 45–51. [Google Scholar] [CrossRef]
  196. Porrini, M.P.; Audisio, M.C.; Sabaté, D.C.; Ibarguren, C.; Medici, S.K.; Sarlo, E.G.; Garrido, P.M.; Eguaras, M.J. Effect of bacterial metabolites on microsporidian Nosema ceranae and on its host Apis mellifera. Parasitol. Res. 2010, 107, 381–388. [Google Scholar] [CrossRef]
  197. Andrearczyk, S.; Kadhim, M.J.; Knaga, S. Influence of a probiotic on the mortality, sugar syrup ingestion and infection of honeybees with Nosema spp. under laboratory assessment. Med. Weter 2014, 70, 762. [Google Scholar]
  198. Borges, D.; Guzman-Novoa, E.; Goodwin, P.H. Effects of prebiotics and probiotics on honey bees (Apis mellifera) infected with the microsporidian parasite Nosema ceranae. Microorganisms 2021, 9, 481. [Google Scholar] [CrossRef] [PubMed]
  199. Ptaszyńska, A.A.; Borsuk, G.; Zdybicka-Barabas, A.; Cytryńska, M.; Małek, W. Are commercial probiotics and prebiotics effective in the treatment and prevention of honeybee nosemosis C? Parasitol. Res. 2016, 115, 397–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. El Khoury, S.; Rousseau, A.; Lecoeur, A.; Cheaib, B.; Bouslama, S.; Mercier, P.-L.; Demey, V.; Castex, M.; Giovenazzo, P.; Derome, N. Deleterious interaction between honeybees (Apis mellifera) and its microsporidian intracellular parasite Nosema ceranae was mitigated by administrating either endogenous or allochthonous gut microbiota strains. Front. Ecol. Evol. 2018, 6, 58. [Google Scholar] [CrossRef] [Green Version]
  201. De Piano, F.G.; Maggi, M.D.; Arceitto, F.R.M.; Audisio, M.C.; Eguaras, M.; Ruffinengo, S.R. Effects of bacterial cell-free supernatant on nutritional parameters of Apis mellifera and their toxicity against Varroa destructor. J. Apic. Sci. 2020, 64, 55–66. [Google Scholar] [CrossRef]
  202. Saccà, M.; Lodesani, M. Isolation of bacterial microbiota associated to honey bees and evaluation of potential biocontrol agents of Varroa destructor. Benef. Microbes 2020, 11, 641–654. [Google Scholar] [CrossRef]
  203. Khongphinitbunjong, K.; de Guzman, L.I.; Tarver, M.R.; Rinderer, T.E.; Chen, Y.; Chantawannakul, P. Differential viral levels and immune gene expression in three stocks of Apis mellifera induced by different numbers of Varroa destructor. J. Insect Physiol. 2015, 72, 28–34. [Google Scholar] [CrossRef] [PubMed]
  204. Antúnez, K.; Martín-Hernández, R.; Prieto, L.; Meana, A.; Zunino, P.; Higes, M. Immune suppression in the honey bee (Apis mellifera) following infection by Nosema ceranae (Microsporidia). Environ. Microbiol. 2009, 11, 2284–2290. [Google Scholar] [CrossRef]
  205. Li, W.; Chen, Y.; Cook, S.C. Chronic Nosema ceranae infection inflicts comprehensive and persistent immunosuppression and accelerated lipid loss in host Apis mellifera honey bees. Int. J. Parasitol. 2018, 48, 433–444. [Google Scholar] [CrossRef] [PubMed]
  206. Roberts, K.E.; Hughes, W.O.H. Immunosenescence and resistance to parasite infection in the honey bee, Apis mellifera. J. Invertebr. Pathol. 2014, 121, 1–6. [Google Scholar] [CrossRef] [PubMed]
  207. Gregorc, A.; Evans, J.D.; Scharf, M.; Ellis, J.D. Gene expression in honey bee (Apis mellifera) larvae exposed to pesticides and Varroa mites (Varroa destructor). J. Insect Physiol. 2012, 58, 1042–1049. [Google Scholar] [CrossRef] [PubMed]
  208. Li, G.; Zhao, H.; Liu, Z.; Wang, H.; Xu, B.; Guo, X. The wisdom of honeybee defenses against environmental stresses. Front. Microbiol. 2018, 9, 722. [Google Scholar] [CrossRef] [PubMed]
  209. Larsen, A.; Reynaldi, F.J.; Guzmán-Novoa, E. Fundaments of the honey bee (Apis mellifera) immune system. Review. Rev. Mex. Cienc. Pecu. 2019, 10, 705–728. [Google Scholar] [CrossRef]
  210. Yi, H.-Y.; Chowdhury, M.; Huang, Y.-D.; Yu, X.-Q. Insect antimicrobial peptides and their applications. Appl. Microbiol. Biotechnol. 2014, 98, 5807–5822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Wu, Q.; Patočka, J.; Kuča, K. Insect antimicrobial peptides, a mini review. Toxins 2018, 10, 461. [Google Scholar] [CrossRef] [PubMed]
  212. Kwong, W.K.; Mancenido, A.L.; Moran, N.A. Immune system stimulation by the native gut microbiota of honey bees. R. Soc. Open Sci. 2017, 4, 170003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Emery, O.; Schmidt, K.; Engel, P. Immune system stimulation by the gut symbiont Frischella perrara in the honey bee (Apis mellifera). Mol. Ecol. 2017, 26, 2576–2590. [Google Scholar] [CrossRef] [PubMed]
  214. Brown, M. Modes of action of probiotics: Recent developments. J. Anim. Vet. Adv. 2011, 10, 1895–1900. [Google Scholar] [CrossRef]
  215. Behnsen, J.; Raffatellu, M. Siderophores: More than stealing iron. MBio 2016, 7, e01906-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Sorrentino, E.; Tremonte, P.; Succi, M.; Iorizzo, M.; Pannella, G.; Lombardi, S.J.; Sturchio, M.; Coppola, R. Detection of antilisterial activity of 3-Phenyllactic acid using Listeria innocua as a model. Front. Microbiol. 2018, 9, 1373. [Google Scholar] [CrossRef]
  217. Kleerebezem, M.; Hols, P.; Bernard, E.; Rolain, T.; Zhou, M.; Siezen, R.J.; Bron, P.A. The extracellular biology of the lactobacilli. FEMS Microbiol. Rev. 2010, 34, 199–230. [Google Scholar] [CrossRef] [PubMed]
  218. Alakomi, H.L.; Skyttä, E.; Saarela, M.; Mattila-Sandholm, T.; Latva-Kala, K.; Helander, I.M. Lactic acid permeabilizes gram-negative bacteria by disrupting the outer membrane. Appl. Environ. Microbiol. 2000, 66, 2001–2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Hassan, M.; Kjos, M.; Nes, I.F.; Diep, D.B.; Lotfipour, F. Natural antimicrobial peptides from bacteria: Characteristics and potential applications to fight against antibiotic resistance. J. Appl. Microbiol. 2012, 113, 723–736. [Google Scholar] [CrossRef]
  220. Reis, J.; Paula, A.; Casarotti, S.; Penna, A. Lactic acid bacteria antimicrobial compounds: Characteristics and applications. Food Eng. Rev. 2012, 4, 124–140. [Google Scholar] [CrossRef]
  221. Yang, S.-C.; Lin, C.-H.; Sung, C.T.; Fang, J.-Y. Antibacterial activities of bacteriocins: Application in foods and pharmaceuticals. Front. Microbiol. 2014, 5, 241. [Google Scholar] [PubMed] [Green Version]
  222. Ovchinnikov, K.V.; Chi, H.; Mehmeti, I.; Holo, H.; Nes, I.F.; Diep, D.B. Novel group of leaderless multipeptide bacteriocins from gram-positive bacteria. Appl. Environ. Microbiol. 2016, 82, 5216–5224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Simons, A.; Alhanout, K.; Duval, R.E. Bacteriocins, antimicrobial peptides from bacterial origin: Overview of their biology and their impact against multidrug-resistant bacteria. Microorganisms 2020, 8, 639. [Google Scholar] [CrossRef] [PubMed]
  224. Janashia, I.; Choiset, Y.; Rabesona, H.; Hwanhlem, N.; Bakuradze, N.; Chanishvili, N.; Haertlé, T. Protection of honeybee Apis mellifera by its endogenous and exogenous lactic flora against bacterial infections. Ann. Agrar. Sci. 2016, 14, 177–181. [Google Scholar] [CrossRef]
  225. Aronstein, K.A.; Murray, K.D. Chalkbrood disease in honey bees. J. Invertebr. Pathol. 2010, 103, S20–S29. [Google Scholar] [CrossRef] [PubMed]
  226. Albo, G.N.; Cordoba, S.B.; Reynaldi, F.J. Chalkbrood: Pathogenesis and the interaction with honeybee defenses. Int. J. Environ. Agric. Res. 2017, 3, 71–80. [Google Scholar]
  227. Kluser, S.; Peduzzi, P. Global Pollinator Decline: A Literature Review; UNEP: Geneva, Switzerland, 2007. [Google Scholar]
  228. Zaghloul, O.; Mourad, A.; El Kady, M.B.; Nemat, F.; Morsy, M. Assessment of losses in honey yield due to the chalkbrood disease, with reference to the determination of its economic injury levels in Egypt. Commun. Agric. Appl. Biol. Sci. 2005, 70, 703–714. [Google Scholar] [PubMed]
  229. Vojvodic, S.; Boomsma, J.J.; Eilenberg, J.; Jensen, A.B. Virulence of mixed fungal infections in honey bee brood. Front. Zool. 2012, 9, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Evison, S.E. Chalkbrood: Epidemiological perspectives from the host–parasite relationship. Curr. Opin. Insect Sci. 2015, 10, 65–70. [Google Scholar] [CrossRef] [PubMed]
  231. Heath, L.A.F. Development of chalk brood in a honeybee colony: A review. Bee World 1982, 63, 119–130. [Google Scholar] [CrossRef]
  232. Liu, T. Ultrastructural changes in the spore and mycelia of Ascosphaera apis after treatment with benomyl (Benlate 50 W). Mycopathologia 1991, 116, 23–28. [Google Scholar] [CrossRef]
  233. Hornitzky, M. Literature Review of Chalkbrood. A Report for the RIRDC; Publication No. 01/150; NSW Agriculture: Kingston, ACT, Australia, 2001.
  234. Davis, C. Control of Chalkbrood Disease with Natural Products: A Report for the Rural Industries Research and Development Corporation; RIRDC publication no. 03/107; Davis, C., Ward, W., Eds.; Rural Industries Research and Development Corporation: Barton, ACT, Australia, 2003.
  235. Kostecka, M.; Niewiadomy, A. Antifungal activity of new series of compound against Ascosphaera apis. Mikol. Lek 2010, 17, 169–171. [Google Scholar]
  236. Strachecka, A.; Borsuk, G.; Olszewski, K.; Paleolog, J.; Gagos, M.; Chobotow, J.; Nawrocka, A.; Gryzinska, M.; Bajda, M. The effect of amphotericin B on the lifespan, body-surface protein concentrations, and DNA methylation levels of honey bees (Apis mellifera). J. Apic. Sci. 2012, 56, 107. [Google Scholar] [CrossRef] [Green Version]
  237. Al-Waili, N.; Salom, K.; Al-Ghamdi, A.; Ansari, M.J. Antibiotic, pesticide, and microbial contaminants of honey: Human health hazards. Sci. World J. 2012, 2012, 930849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. McFrederick, Q.S.; Mueller, U.G.; James, R.R. Interactions between fungi and bacteria influence microbial community structure in the Megachile rotundata larval gut. Proc. Biol. Sci. 2014, 281, 20132653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Sellamuthu, P.S.; Sivakumar, D.; Soundy, P. Antifungal activity and chemical composition of thyme, peppermint and citronella oils in vapor phase against avocado and peach postharvest pathogens. J. Food Saf. 2013, 33, 86–93. [Google Scholar] [CrossRef]
  240. Ansari, M.J.; Al-Ghamdi, A.; Usmani, S.; Khan, K.A.; Alqarni, A.S.; Kaur, M.; Al-Waili, N. In vitro evaluation of the effects of some plant essential oils on Ascosphaera apis, the causative agent of chalkbrood disease. Curr. Res. Apic. 2017, 24, 1001–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Gabriel, K.T.; Kartforosh, L.; Crow, S.A.; Cornelison, C.T. Antimicrobial activity of essential oils against the fungal pathogens Ascosphaera apis and Pseudogymnoascus destructans. Mycopathologia 2018, 183, 921–934. [Google Scholar] [CrossRef] [PubMed]
  242. Nardoni, S.; D’Ascenzi, C.; Rocchigiani, G.; Papini, R.A.; Pistelli, L.; Formato, G.; Najar, B.; Mancianti, F. Stonebrood and chalkbrood in Apis mellifera causing fungi: In vitro sensitivity to some essential oils. Nat. Prod. Res. 2018, 32, 385–390. [Google Scholar] [CrossRef] [PubMed]
  243. Simone-Finstrom, M.D.; Spivak, M. Increased resin collection after parasite challenge: A case of self-medication in honey bees? PLoS ONE 2012, 7, e34601. [Google Scholar] [CrossRef] [Green Version]
  244. Voigt, K.; Rademacher, E. Effect of the propolis components, cinnamic acid and pinocembrin, on Apis mellifera and Ascosphaera apis. J. Apic. Sci. 2015, 59, 89–95. [Google Scholar] [CrossRef] [Green Version]
  245. Wilson, M.B.; Brinkman, D.; Spivak, M.; Gardner, G.; Cohen, J.D. Regional variation in composition and antimicrobial activity of US propolis against PaeniBacillus larvae and Ascosphaera apis. J. Invertebr. Pathol. 2015, 124, 44–50. [Google Scholar] [CrossRef] [PubMed]
  246. Simone-Finstrom, M.; Borba, R.S.; Wilson, M.; Spivak, M. Propolis counteracts some threats to honey bee health. Insects 2017, 8, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Reynaldi, F.; De Giusti, M.; Alippi, A. Inhibition of the growth of Ascosphaera apis by Bacillus and Paenibacillus strains isolated from honey. Rev. Argent. Microbiol. 2004, 36, 52–55. [Google Scholar] [PubMed]
  248. Sabaté, D.C.; Carrillo, L.; Carina Audisio, M. Inhibition of PaeniBacillus larvae and Ascosphaera apis by Bacillus subtilis isolated from honeybee gut and honey samples. Res. Microbiol. 2009, 160, 193–199. [Google Scholar] [CrossRef] [PubMed]
  249. Omar, M.O.; Moustafa, A.M.; Ansari, M.J.; Anwar, A.M.; Fahmy, B.F.; Al-Ghamdi, A.; Nuru, A. Antagonistic effect of gut bacteria in the hybrid carniolan honey bee, Apis mellifera carnica, against Ascosphaera apis, the causal organism of chalkbrood disease. J. Apic. Sci. 2014, 58, 17. [Google Scholar] [CrossRef] [Green Version]
  250. Alonso-Salces, R.M.; Cugnata, N.M.; Guaspari, E.; Pellegrini, M.C.; Aubone, I.; De Piano, F.G.; Antunez, K.; Fuselli, S.R. Natural strategies for the control of PaeniBacillus larvae, the causative agent of American foulbrood in honey bees: A review. Apidologie 2017, 48, 387–400. [Google Scholar] [CrossRef]
  251. Poppinga, L.; Genersch, E. Molecular pathogenesis of american foulbrood: How PaeniBacillus larvae kills honey bee larvae. Curr. Opin. Insect Sci. 2015, 10, 29–36. [Google Scholar] [CrossRef] [PubMed]
  252. Müller, S.; Garcia-Gonzalez, E.; Genersch, E.; Süssmuth, R.D. Involvement of secondary metabolites in the pathogenesis of the american foulbrood of honey bees caused by PaeniBacillus larvae. Nat. Prod. Rep. 2015, 32, 765–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Garcia-Gonzalez, E.; Poppinga, L.; Fünfhaus, A.; Hertlein, G.; Hedtke, K.; Jakubowska, A.; Genersch, E. PaeniBacillus larvae chitin-degrading protein Pl CBP49 is a key virulence factor in American foulbrood of honey bees. PLoS Pathog. 2014, 10, e1004284. [Google Scholar] [CrossRef] [PubMed]
  254. Djukic, M.; Brzuszkiewicz, E.; Fünfhaus, A.; Voss, J.; Gollnow, K.; Poppinga, L.; Liesegang, H.; Garcia-Gonzalez, E.; Genersch, E.; Daniel, R. How to kill the honey bee larva: Genomic potential and virulence mechanisms of PaeniBacillus larvae. PLoS ONE 2014, 9, e90914. [Google Scholar]
  255. Hansen, H.; Brødsgaard, C.J. American foulbrood: A review of its biology, diagnosis and control. Bee World 1999, 80, 5–23. [Google Scholar] [CrossRef]
  256. Lindström, A.; Korpela, S.; Fries, I. The distribution of PaeniBacillus larvae spores in adult bees and honey and larval mortality, following the addition of American foulbrood diseased brood or spore-contaminated honey in honey bee (Apis mellifera) colonies. J. Invertebr. Pathol. 2008, 99, 82–86. [Google Scholar] [CrossRef] [PubMed]
  257. Fries, I.; Camazine, S. Implications of horizontal and vertical pathogen transmission for honey bee epidemiology. Apidologie 2001, 32, 199–214. [Google Scholar] [CrossRef] [Green Version]
  258. Genersch, E. PaeniBacillus larvae and American foulbrood–Long since known and still surprising. J. Verbrauch. Lebensm. 2008, 3, 429–434. [Google Scholar] [CrossRef]
  259. Datta, S.; Bull, J.C.; Budge, G.E.; Keeling, M.J. Modelling the spread of American foulbrood in honeybees. J. R. Soc. Interface 2013, 10, 20130650. [Google Scholar] [CrossRef] [PubMed]
  260. Forsgren, E.; Stevanovic, J.; Fries, I. Variability in germination and in temperature and storage resistance among PaeniBacillus larvae genotypes. Vet. Microbiol. 2008, 129, 342–349. [Google Scholar] [CrossRef]
  261. Bailey, L.; Collins, M. Reclassification of ‘Streptococcus pluton’(white) in a new genus Melissococcus, as Melissococcus pluton nom. rev.; comb. nov. J. Appl. Bacteriol. 1982, 53, 215–217. [Google Scholar] [CrossRef]
  262. Ellis, J.D.; Munn, P.A. The worldwide health status of honey bees. Bee World 2005, 86, 88–101. [Google Scholar] [CrossRef]
  263. Lewkowski, O.; Erler, S. Virulence of Melissococcus plutonius and secondary invaders associated with European foulbrood disease of the honey bee. MicrobiologyOpen 2019, 8, e00649. [Google Scholar] [CrossRef] [PubMed]
  264. Bailey, L. Melissococcus pluton, the cause of European foulbrood of honey bees (Apis Spp.). J. Appl. Bacteriol. 1983, 55, 65–69. [Google Scholar] [CrossRef]
  265. Bailey, L.; Ball, B.V. 4—Bacteria. In Honey Bee Pathology, 2nd ed.; Bailey, L., Ball, B.V., Eds.; Academic Press: London, UK, 1991; pp. 35–52. ISBN 978-0-12-073481-8. [Google Scholar]
  266. Bailey, L. Recent research on the natural history of European foul brood disease. Bee World 1959, 40, 66–70. [Google Scholar] [CrossRef]
  267. Belloy, L.; Imdorf, A.; Fries, I.; Forsgren, E.; Berthoud, H.; Kuhn, R.; Charrière, J.-D. Spatial distribution of Melissococcus plutonius in adult honey bees collected from apiaries and colonies with and without symptoms of European foulbrood. Apidologie 2007, 38, 136–140. [Google Scholar] [CrossRef]
  268. Sopko, B.; Zitek, J.; Nesvorna, M.; Markovic, M.; Kamler, M.; Titera, D.; Erban, T.; Hubert, J. Detection and quantification of Melissococcus plutonius in honey bee workers exposed to European foulbrood in Czechia through conventional PCR, QPCR, and barcode sequencing. J. Apic. Res. 2020, 59, 503–514. [Google Scholar] [CrossRef]
  269. Forsgren, E.; Locke, B.; Sircoulomb, F.; Schäfer, M.O. Bacterial Diseases in Honeybees. Curr. Clin. Microbiol. Rep. 2018, 5, 18–25. [Google Scholar] [CrossRef]
  270. Wakgari, M.; Yigezu, G. Honeybee Keeping Constraints and Future Prospects. Cogent Food Agric. 2021, 7, 1872192. [Google Scholar] [CrossRef]
  271. Thompson, H.M.; Brown, M.A. Is contact colony treatment with antibiotics an effective control for European foulbrood? Bee World 2001, 82, 130–138. [Google Scholar] [CrossRef]
  272. Evans, J.D. Diverse origins of tetracycline resistance in the honey bee bacterial pathogen PaeniBacillus larvae. J. Invertebr. Pathol. 2003, 83, 46–50. [Google Scholar] [CrossRef]
  273. Miyagi, T.; Peng, C.Y.S.; Chuang, R.Y.; Mussen, E.C.; Spivak, M.S.; Doi, R.H. Verification of oxytetracycline-resistant american foulbrood pathogen PaeniBacillus larvae in the United States. J. Invertebr. Pathol. 2000, 75, 95–96. [Google Scholar] [CrossRef] [PubMed]
  274. Kochansky, J.; Knox, D.A.; Feldlaufer, M.; Pettis, J.S. Screening alternative antibiotics against oxytetracycline-susceptible and-resistant PaeniBacillus larvae. Apidologie 2001, 32, 215–222. [Google Scholar] [CrossRef] [Green Version]
  275. Alippi, A.M.; Leon, I.E.; López, A.C. Tetracycline-resistance encoding plasmids from PaeniBacillus larvae, the causal agent of American foulbrood disease, isolated from commercial honeys. Int. Microbiol. 2014, 17, 49–61. [Google Scholar] [PubMed]
  276. Krongdang, S.; Evans, J.D.; Pettis, J.S.; Chantawannakul, P. Multilocus sequence typing, biochemical and antibiotic resistance characterizations reveal diversity of North American strains of the honey bee pathogen PaeniBacillus larvae. PLoS ONE 2017, 12, e0176831. [Google Scholar] [CrossRef] [PubMed]
  277. Erban, T.; Ledvinka, O.; Kamler, M.; Nesvorna, M.; Hortova, B.; Tyl, J.; Titera, D.; Markovic, M.; Hubert, J. Honeybee (Apis mellifera)-associated bacterial community affected by american foulbrood: Detection of PaeniBacillus larvae via microbiome analysis. Sci. Rep. 2017, 7, 5084. [Google Scholar] [CrossRef] [PubMed]
  278. Alberoni, D.; Gaggìa, F.; Baffoni, L.; Di Gioia, D. Beneficial microorganisms for honey bees: Problems and progresses. Appl. Microbiol. Biotechnol. 2016, 100, 9469–9482. [Google Scholar] [CrossRef] [PubMed]
  279. Bargańska, Ż.; Namieśnik, J.; Ślebioda, M. Determination of antibiotic residues in honey. TrAC Trends Anal. Chem. 2011, 30, 1035–1041. [Google Scholar] [CrossRef]
  280. Lopez, M.I.; Pettis, J.S.; Smith, I.B.; Chu, P.-S. Multiclass determination and confirmation of antibiotic residues in honey using LC-MS/MS. J. Agric. Food Chem. 2008, 56, 1553–1559. [Google Scholar] [CrossRef]
  281. Floyd, A.S.; Mott, B.M.; Maes, P.; Copeland, D.C.; McFrederick, Q.S.; Anderson, K.E. Microbial ecology of European foul brood disease in the honey bee (Apis mellifera): Towards a microbiome understanding of disease susceptibility. Insects 2020, 11, 555. [Google Scholar] [CrossRef] [PubMed]
  282. Al-Ghamdi, A.A.; Al-Ghamdi, M.S.; Ahmed, A.M.; Mohamed, A.S.A.; Shaker, G.H.; Ansari, M.J.; Dorrah, M.A.; Khan, K.A.; Ayaad, T.H. Immune investigation of the honeybee Apis mellifera jemenitica broods: A step toward production of a bee-derived antibiotic against the American foulbrood. Saudi J. Biol. Sci. 2021, 28, 1528–1538. [Google Scholar] [CrossRef] [PubMed]
  283. Grady, E.N.; MacDonald, J.; Liu, L.; Richman, A.; Yuan, Z.-C. Current knowledge and perspectives of paenibacillus: A review. Microb. Cell Factories 2016, 15, 203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Antúnez, K.; Harriet, J.; Gende, L.; Maggi, M.; Eguaras, M.; Zunino, P. Efficacy of natural propolis extract in the control of american foulbrood. Vet. Microbiol. 2008, 131, 324–331. [Google Scholar] [CrossRef] [PubMed]
  285. Flesar, J.; Havlik, J.; Kloucek, P.; Rada, V.; Titera, D.; Bednar, M.; Stropnicky, M.; Kokoska, L. In vitro growth-inhibitory effect of plant-derived extracts and compounds against PaeniBacillus larvae and their acute oral toxicity to adult honey bees. Vet. Microbiol. 2010, 145, 129–133. [Google Scholar] [CrossRef] [PubMed]
  286. Santos, R.C.V.; Lopes, L.Q.S.; dos Alves, C.F.S.; Fausto, V.P.; Pizzutti, K.; Barboza, V.; de Souza, M.E.; Raffin, R.P.; Gomes, P.; Takamatsu, D.; et al. Antimicrobial activity of tea tree oil nanoparticles against American and European foulbrood diseases agents. J. Asia-Pac. Entomol. 2014, 17, 343–347. [Google Scholar] [CrossRef]
  287. Santos, R.C.V.; dos Alves, C.F.S.; Schneider, T.; Lopes, L.Q.S.; Aurich, C.; Giongo, J.L.; Brandelli, A.; de Almeida Vaucher, R. Antimicrobial activity of Amazonian oils against Paenibacillus species. J. Invertebr. Pathol. 2012, 109, 265–268. [Google Scholar] [CrossRef]
  288. Chaimanee, V.; Thongtue, U.; Sornmai, N.; Songsri, S.; Pettis, J.S. Antimicrobial activity of plant extracts against the honeybee pathogens, PaeniBacillus larvae and Ascosphaera apis and their topical toxicity to Apis mellifera adults. J. Appl. Microbiol. 2017, 123, 1160–1167. [Google Scholar] [CrossRef] [PubMed]
  289. Kim, J.; Park, S.; Shin, Y.-K.; Kang, H.; Kim, K.-Y. In vitro antibacterial activity of macelignan and corosolic acid against the bacterial bee pathogens PaeniBacillus larvae and Melissococcus plutonius. Acta Vet. Brno 2018, 87, 277–284. [Google Scholar] [CrossRef]
  290. Fuselli, S.R.; de la Rosa, S.B.G.; Eguaras, M.J.; Fritz, R. Chemical composition and antimicrobial activity of citrus essences on honeybee bacterial pathogen PaeniBacillus larvae, the causal agent of American foulbrood. World J. Microbiol. Biotechnol. 2008, 24, 2067–2072. [Google Scholar] [CrossRef]
  291. Bilikova, K.; Popova, M.; Trusheva, B.; Bankova, V. New anti-PaeniBacillus larvae substances purified from propolis. Apidologie 2013, 44, 278–285. [Google Scholar] [CrossRef] [Green Version]
  292. Isidorov, V.A.; Buczek, K.; Zambrowski, G.; Miastkowski, K.; Swiecicka, I. In vitro study of the antimicrobial activity of European propolis against PaeniBacillus larvae. Apidologie 2017, 48, 411–422. [Google Scholar] [CrossRef] [Green Version]
  293. Chen, Y.-W.; Ye, S.-R.; Ting, C.; Yu, Y.-H. Antibacterial activity of propolins from Taiwanese green propolis. J. Food Drug Anal. 2018, 26, 761–768. [Google Scholar] [CrossRef] [Green Version]
  294. Borba, R.S.; Spivak, M. Propolis envelope in Apis mellifera colonies supports honey bees against the pathogen, PaeniBacillus larvae. Sci. Rep. 2017, 7, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Cecotti, R.; Bergomi, P.; Carpana, E.; Tava, A. Chemical characterization of the volatiles of leaves and flowers from cultivated Malva sylvestris var. mauritiana and their antimicrobial activity against the aetiological agents of the European and American foulbrood of honeybees (Apis mellifera). Nat. Prod. Commun. 2016, 11, 1527–1530. [Google Scholar] [CrossRef] [Green Version]
  296. Fernández, N.J.; Damiani, N.; Podaza, E.A.; Martucci, J.F.; Fasce, D.; Quiroz, F.; Meretta, P.E.; Quintana, S.; Eguaras, M.J.; Gende, L.B. Laurus nobilis L. extracts against PaeniBacillus larvae: Antimicrobial activity, antioxidant capacity, hygienic behavior and colony strength. Saudi J. Biol. Sci. 2019, 26, 906–912. [Google Scholar] [CrossRef]
  297. Floris, I.; Carta, C.; Moretti, M. Activity of various essential oils against Bacillus larvae white in vitro and in apiary trials. Apidologie 1996, 27, 111–119. [Google Scholar] [CrossRef] [Green Version]
  298. Gende, L.B.; Floris, I.; Fritz, R.; Eguaras, M.J. Antimicrobial activity of cinnamon (Cinnamomum zeylanicum) essential oil and its main components against PaeniBacillus larvae from Argentine. Bull. Insectol. 2008, 61, 1. [Google Scholar]
  299. Yoshiyama, M.; Kimura, K. Bacteria in the gut of Japanese honeybee, Apis cerana japonica, and their antagonistic effect against PaeniBacillus larvae, the causal agent of American foulbrood. J. Invertebr. Pathol. 2009, 102, 91–96. [Google Scholar] [CrossRef]
  300. Fries, I.; Feng, F.; da Silva, A.; Slemenda, S.B.; Pieniazek, N.J. Nosema ceranae n. sp. (Microspora, Nosematidae), morphological and molecular characterization of a microsporidian parasite of the Asian honey bee Apis cerana (Hymenoptera, Apidae). Eur. J. Protistol. 1996, 32, 356–365. [Google Scholar] [CrossRef]
  301. Adl, S.M.; Simpson, A.G.B.; Farmer, M.A.; Andersen, R.A.; Anderson, O.R.; Barta, J.R.; Bowser, S.S.; Brugerolle, G.; Fensome, R.A.; Fredericq, S.; et al. The new higher level classification of eukaryotes with emphasis on the taxonomy of protists. J. Eukaryot. Microbiol. 2005, 52, 399–451. [Google Scholar] [CrossRef] [PubMed]
  302. Corradi, N.; Keeling, P.J. Microsporidia: A journey through radical taxonomical revisions. Fungal Biol. Rev. 2009, 23, 1–8. [Google Scholar] [CrossRef]
  303. Sulborska, A.; Horecka, B.; Cebrat, M.; Kowalczyk, M.; Skrzypek, T.H.; Kazimierczak, W.; Trytek, M.; Borsuk, G. Microsporidia Nosema Spp. Obligate Bee Parasites Are Transmitted by Air. Sci. Rep. 2019, 9, 14376. [Google Scholar] [CrossRef]
  304. Goblirsch, M. Nosema ceranae disease of the honey bee (Apis mellifera). Apidologie 2018, 49, 131–150. [Google Scholar] [CrossRef] [Green Version]
  305. Paris, L.; El Alaoui, H.; Delbac, F.; Diogon, M. Effects of the gut parasite Nosema ceranae on honey bee physiology and behavior. Curr. Opin. Insect Sci. 2018, 26, 149–154. [Google Scholar] [CrossRef]
  306. Higes, M.; Martín-Hernández, R.; Meana, A. Nosema ceranae in Europe: An emergent type C nosemosis. Apidologie 2010, 41, 375–392. [Google Scholar] [CrossRef] [Green Version]
  307. Fries, I. Nosema apis—A parasite in the honey bee colony. Bee World 1993, 74, 5–19. [Google Scholar] [CrossRef]
  308. Dosselli, R.; Grassl, J.; Carson, A.; Simmons, L.W.; Baer, B. Flight behaviour of honey bee (Apis mellifera) workers is altered by initial infections of the fungal parasite Nosema apis. Sci. Rep. 2016, 6, 36649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  309. Higes, M.; García-Palencia, P.; Martín-Hernández, R.; Meana, A. Experimental infection of Apis mellifera honeybees with Nosema ceranae (Microsporidia). J. Invertebr. Pathol. 2007, 94, 211–217. [Google Scholar] [CrossRef]
  310. Higes, M.; Martín-Hernández, R.; Botías, C.; Bailón, E.G.; González-Porto, A.V.; Barrios, L.; del Nozal, M.J.; Bernal, J.L.; Jiménez, J.J.; Palencia, P.G.; et al. How natural infection by Nosema ceranae causes honeybee colony collapse. Environ. Microbiol. 2008, 10, 2659–2669. [Google Scholar] [CrossRef] [PubMed]
  311. Paxton, R.J. Does Infection by Nosema ceranae cause “colony collapse disorder” in honey bees (Apis mellifera)? J. Apic. Res. 2010, 49, 80–84. [Google Scholar] [CrossRef]
  312. Botías, C.; Martín-Hernández, R.; Barrios, L.; Meana, A.; Higes, M. Nosema spp. infection and its negative effects on honey bees (Apis mellifera iberiensis) at the colony level. Vet. Res. 2013, 44, 25. [Google Scholar] [CrossRef] [Green Version]
  313. Goblirsch, M.; Huang, Z.Y.; Spivak, M. Physiological and behavioral changes in honey bees (Apis mellifera) induced by Nosema ceranae infection. PLoS ONE 2013, 8, e58165. [Google Scholar] [CrossRef] [PubMed]
  314. Aufauvre, J.; Misme-Aucouturier, B.; Viguès, B.; Texier, C.; Delbac, F.; Blot, N. Transcriptome analyses of the honeybee response to Nosema ceranae and insecticides. PLoS ONE 2014, 9, e91686. [Google Scholar] [CrossRef]
  315. Basualdo, M.; Barragán, S.; Antúnez, K. Bee bread increases honeybee haemolymph protein and promote better survival despite of causing higher Nosema ceranae abundance in honeybees. Environ. Microbiol. Rep. 2014, 6, 396–400. [Google Scholar] [CrossRef] [PubMed]
  316. Gage, S.L.; Kramer, C.; Calle, S.; Carroll, M.; Heien, M.; DeGrandi-Hoffman, G. Nosema ceranae parasitism impacts olfactory learning and memory and neurochemistry in honey bees (Apis mellifera). J. Exp. Biol. 2018, 221, jeb161489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. Graystock, P.; Goulson, D.; Hughes, W.O. Parasites in bloom: Flowers aid dispersal and transmission of pollinator parasites within and between bee species. Proc. R. Soc. B Biol. Sci. 2015, 282, 20151371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. European Commission. Commission Regulation (EU) No 37/2010 of 22 December 2009 on Pharmacologically Active Substances and Their Classification Regarding Maximum Residue Limits in Foodstuffs of Animal Origin. J. Eur. Union 2010, 15, 1–72.
  319. Van den Heever, J.P.; Thompson, T.S.; Otto, S.J.G.; Curtis, J.M.; Ibrahim, A.; Pernal, S.F. Evaluation of Fumagilin-B® and other potential alternative chemotherapies against Nosema ceranae-infected honeybees (Apis mellifera) in cage trial assays. Apidologie 2016, 47, 617–630. [Google Scholar] [CrossRef] [Green Version]
  320. Van den Heever, J.P.; Thompson, T.S.; Curtis, J.M.; Ibrahim, A.; Pernal, S.F. Fumagillin: An overview of recent scientific advances and their significance for apiculture. J. Agric. Food Chem. 2014, 62, 2728–2737. [Google Scholar] [CrossRef] [PubMed]
  321. Underwood, R.M.; Currie, R.W. Indoor winter fumigation with formic acid for control of Acarapis woodi (Acari: Tarsonemidae) and nosema disease, Nosema sp. J. Econ. Entomol. 2009, 102, 1729–1736. [Google Scholar] [CrossRef]
  322. Maistrello, L.; Lodesani, M.; Costa, C.; Leonardi, F.; Marani, G.; Caldon, M.; Mutinelli, F.; Granato, A. Screening of natural compounds for the control of nosema disease in honeybees (Apis mellifera). Apidologie 2008, 39, 436–445. [Google Scholar] [CrossRef] [Green Version]
  323. Costa, C.; Lodesani, M.; Maistrello, L. Effect of thymol and resveratrol administered with candy or syrup on the development of Nosema ceranae and on the longevity of honeybees (Apis mellifera L.) in laboratory conditions. Apidologie 2010, 41, 141–150. [Google Scholar] [CrossRef] [Green Version]
  324. van den Heever, J.P.; Thompson, T.S.; Otto, S.J.G.; Curtis, J.M.; Ibrahim, A.; Pernal, S.F. The effect of dicyclohexylamine and fumagillin on Nosema ceranae-infected honey bee (Apis mellifera) mortality in cage trial assays. Apidologie 2016, 47, 663–670. [Google Scholar] [CrossRef]
  325. Nanetti, A.; Rodriguez-García, C.; Meana, A.; Martín-Hernández, R.; Higes, M. Effect of oxalic acid on Nosema ceranae infection. Res. Vet. Sci. 2015, 102, 167–172. [Google Scholar] [CrossRef] [PubMed]
  326. Porrini, M.; Garrido, P.; Silva, J.; Cuniolo, A.; Román, S.; Iaconis, D.; Eguaras, M. Ácido oxálico: Potencial antiparasitario frente a Nosema ceranae por administración oral y exposición total. In Proceedings of the I Workshop Latinoamericano en Sanidad Apicola, Mar de Plata, Argentina, 25–26 October 2018; pp. 25–26. [Google Scholar]
  327. Cilia, G.; Garrido, C.; Bonetto, M.; Tesoriero, D.; Nanetti, A. Effect of Api-Bioxal® and ApiHerb® treatments against Nosema ceranae infection in Apis mellifera investigated by two qpcr methods. Vet. Sci. 2020, 7, 125. [Google Scholar] [CrossRef] [PubMed]
  328. Porrini, M.P.; Fernández, N.J.; Garrido, P.M.; Gende, L.B.; Medici, S.K.; Eguaras, M.J. In vivo evaluation of antiparasitic activity of plant extracts on Nosema ceranae (Microsporidia). Apidologie 2011, 42, 700–707. [Google Scholar] [CrossRef] [Green Version]
  329. Damiani, N.; Fernández, N.J.; Porrini, M.P.; Gende, L.B.; Álvarez, E.; Buffa, F.; Brasesco, C.; Maggi, M.D.; Marcangeli, J.A.; Eguaras, M.J. Laurel leaf extracts for honeybee pest and disease management: Antimicrobial, microsporicidal, and acaricidal activity. Parasitol. Res. 2014, 113, 701–709. [Google Scholar] [CrossRef] [PubMed]
  330. Bravo, J.; Carbonell, V.; Sepúlveda, B.; Delporte, C.; Valdovinos, C.E.; Martín-Hernández, R.; Higes, M. Antifungal activity of the essential oil obtained from Cryptocarya alba against infection in honey bees by Nosema ceranae. J. Invertebr. Pathol. 2017, 149, 141–147. [Google Scholar] [CrossRef]
  331. Arismendi, N.; Vargas, M.; López, M.D.; Barría, Y.; Zapata, N. Promising antimicrobial activity against the honey bee parasite Nosema ceranae by methanolic extracts from Chilean native plants and propolis. J. Apic. Res. 2018, 57, 522–535. [Google Scholar] [CrossRef]
  332. Lee, J.; Kim, J.; Jo, M.; Rangachari, B.; Park, J. Anti-nosemosis activity of aster scaber and artemisia dubia aqueous extracts. J. Apic. Sci. 2018, 62, 27–38. [Google Scholar] [CrossRef] [Green Version]
  333. Glavinic, U.; Stevanovic, J.; Ristanic, M.; Rajkovic, M.; Davitkov, D.; Lakic, N.; Stanimirovic, Z. Potential of fumagillin and agaricus blazei mushroom extract to reduce Nosema ceranae in honey bees. Insects 2021, 12, 282. [Google Scholar] [CrossRef]
  334. Saltykova, E.; Gaifullina, L.; Kaskinova, M.; Gataullin, A.; Matniyazov, R.; Poskryakov, A.; Nikolenko, A. Effect of chitosan on development of Nosema apis microsporidia in honey bees. Microbiology 2018, 87, 738–743. [Google Scholar] [CrossRef]
  335. Valizadeh, P.; Guzman-Novoa, E.; Goodwin, P.H. Effect of immune inducers on Nosema ceranae multiplication and their impact on honey bee (Apis mellifera L.) survivorship and behaviors. Insects 2020, 11, 572. [Google Scholar] [CrossRef] [PubMed]
  336. Nanetti, A.; Ugolini, L.; Cilia, G.; Pagnotta, E.; Malaguti, L.; Cardaio, I.; Matteo, R.; Lazzeri, L. Seed meals from brassica nigra and eruca sativa control artificial Nosema ceranae infections in Apis mellifera. Microorganisms 2021, 9, 949. [Google Scholar] [CrossRef] [PubMed]
  337. Ugolini, L.; Cilia, G.; Pagnotta, E.; Malaguti, L.; Capano, V.; Guerra, I.; Zavatta, L.; Albertazzi, S.; Matteo, R.; Lazzeri, L.; et al. Glucosinolate bioactivation by Apis mellifera workers and its impact on Nosema ceranae infection at the colony level. Biomolecules 2021, 11, 1657. [Google Scholar] [CrossRef] [PubMed]
  338. Borges, D.; Guzman-Novoa, E.; Goodwin, P.H. Control of the microsporidian parasite Nosema ceranae in honey bees (Apis mellifera) using nutraceutical and immuno-stimulatory compounds. PLoS ONE 2020, 15, e0227484. [Google Scholar] [CrossRef] [PubMed]
  339. Corby-Harris, V.; Snyder, L.; Meador, C.; Naldo, R.; Mott, B.; Anderson, K. Parasaccharibacter Apium, gen. nov., sp. nov., improves honey bee (Hymenoptera: Apidae) resistance to Nosema. J. Econ. Entomol. 2016, 109, 537–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  340. Ptaszyńska, A.A.; Borsuk, G.; Mułenko, W.; Wilk, J. Impact of vertebrate probiotics on honeybee yeast microbiota and on the course of nosemosis. Med. Weter. 2016, 72, 430–434. [Google Scholar] [CrossRef]
  341. Dainat, B.; Evans, J.D.; Chen, Y.P.; Gauthier, L.; Neumann, P. Predictive markers of honey bee colony collapse. PLoS ONE 2012, 7, e32151. [Google Scholar] [CrossRef] [PubMed]
  342. Ramsey, S.D.; Ochoa, R.; Bauchan, G.; Gulbronson, C.; Mowery, J.D.; Cohen, A.; Lim, D.; Joklik, J.; Cicero, J.M.; Ellis, J.D. Varroa destructor feeds primarily on honey bee fat body tissue and not hemolymph. Proc. Natl. Acad. Sci. USA 2019, 116, 1792–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Chen, Y.; Pettis, J.S.; Evans, J.D.; Kramer, M.; Feldlaufer, M.F. Transmission of kashmir bee virus by the ectoparasitic mite Varroa destructor. Apidologie 2004, 35, 441–448. [Google Scholar] [CrossRef] [Green Version]
  344. Lanzi, G.; De Miranda, J.R.; Boniotti, M.B.; Cameron, C.E.; Lavazza, A.; Capucci, L.; Camazine, S.M.; Rossi, C. Molecular and biological characterization of deformed wing virus of honeybees (Apis mellifera L.). J. Virol. 2006, 80, 4998–5009. [Google Scholar] [CrossRef] [Green Version]
  345. Burritt, N.L.; Foss, N.J.; Neeno-Eckwall, E.C.; Church, J.O.; Hilger, A.M.; Hildebrand, J.A.; Warshauer, D.M.; Perna, N.T.; Burritt, J.B. Sepsis and hemocyte loss in honey bees (Apis mellifera) infected with Serratia marcescens strain sicaria. PLoS ONE 2016, 11, e0167752. [Google Scholar] [CrossRef] [Green Version]
  346. Ramsey, S.; Gulbronson, C.J.; Mowery, J.; Ochoa, R.; Bauchan, G. A Multi-microscopy approach to discover the feeding site and host tissue consumed by Varroa destructor on host honey bees. Microsc. Microanal. 2018, 24, 1258–1259. [Google Scholar] [CrossRef] [Green Version]
  347. Amdam, G.V.; Hartfelder, K.; Norberg, K.; Hagen, A.; Omholt, S.W. Altered physiology in worker honey bees (Hymenoptera: Apidae) infested with the mite Varroa destructor (Acari: Varroidae): A factor in colony loss during overwintering? J. Econ. Entomol. 2004, 97, 741–747. [Google Scholar] [CrossRef]
  348. Hubert, J.; Kamler, M.; Nesvorna, M.; Ledvinka, O.; Kopecky, J.; Erban, T. Comparison of Varroa destructor and worker honeybee microbiota within hives indicates shared bacteria. Microb. Ecol. 2016, 72, 448–459. [Google Scholar] [CrossRef]
  349. Hubert, J.; Bicianova, M.; Ledvinka, O.; Kamler, M.; Lester, P.J.; Nesvorna, M.; Kopecky, J.; Erban, T. Changes in the bacteriome of honey bees associated with the parasite Varroa destructor, and pathogens Nosema and Lotmaria Passim. Microb. Ecol. 2017, 73, 685–698. [Google Scholar] [CrossRef] [PubMed]
  350. Balakrishnan, B.; Wu, H.; Cao, L.; Zhang, Y.; Li, W.; Han, R. Immune response and hemolymph microbiota of Apis mellifera and Apis cerana after the challenge with recombinant Varroa toxic protein. J. Econ. Entomol. 2021, 114, 1310–1320. [Google Scholar] [CrossRef] [PubMed]
  351. Zhang, Y.; Liu, X.; Zhang, W.; Han, R. Differential gene expression of the honey bees Apis mellifera and A. cerana induced by Varroa destructor infection. J. Insect Physiol. 2010, 56, 1207–1218. [Google Scholar] [CrossRef] [PubMed]
  352. Gracia, M.J.; Moreno, C.; Ferrer, M.; Sanz, A.; Peribáñez, M.Á.; Estrada, R. Field efficacy of acaricides against Varroa destructor. PLoS ONE 2017, 12, e0171633. [Google Scholar] [CrossRef] [PubMed]
  353. Tihelka, E. Effects of synthetic and organic acaricides on honey bee health: A review. Slov. Vet. Res. 2018, 55, 119–140. [Google Scholar] [CrossRef] [Green Version]
  354. Ariana, A.; Ebadi, R.; Tahmasebi, G. Laboratory evaluation of some plant essences to control Varroa destructor (Acari: Varroidae). Exp. Appl. Acarol. 2002, 27, 319–327. [Google Scholar] [CrossRef] [PubMed]
  355. Masry, S.H.D.; Abd El-Wahab, T.E.; Rashad, M. Evaluating the impact of jatropha oil extract against the Varroa Mite, Varroa destructor Anderson & Trueman (Arachnida: Acari: Varroidae), infesting honeybee colonies (Apis mellifera L.). Egypt. J. Biol. Pest Control 2020, 30, 91. [Google Scholar] [CrossRef]
  356. Stanimirović, Z.; Glavinić, U.; Lakić, N.; Radović, D.; Ristanić, M.; Tarić, E.; Stevanović, J. Efficacy of plant-derived formulation argus ras in Varroa destructor control. Acta Vet. 2017, 67, 191–200. [Google Scholar] [CrossRef] [Green Version]
  357. Abd El-Wahab, T.E.; Ebadah, I.; Zidan, E. Control of Varroa mite by essential oils and formic acid with their effects on grooming behaviour of honey bee colonies. J. Basic Appl. Sci. Res. 2012, 2, 7674–7680. [Google Scholar]
  358. Islam, N.; Amjad, M.; Ehsan-ul-Haq, S.E.; Naz, F. Management of Varroa destructor by essential oils and formic acid in Apis mellifera Linn. colonies. J. Entomol. Zool. Stud. 2016, 4, 97–104. [Google Scholar]
  359. Nowar, E.; Khattab, M.; Omar, R.; Mashaal Toka, F. Evaluation of some natural components for controlling Varroa mites in honeybee colonies. Middle East J. Agric. Res. 2018, 7, 264–268. [Google Scholar]
  360. Dimetry, N.; El-Wahab, T.A.; Zakaria, M. Effective control of Varroa mite Varroa destructor Anderson & Trueman infesting honey bee colonies Apis mellifera L. by some natural products. Bull. Fac. Agric. Cairo Univ. 2005, 56, 295–308. [Google Scholar]
  361. Chandler, D.; Sunderland, K.; Ball, B.; Davidson, G. Prospective biological control agents of Varroa destructor n. sp., an important pest of the european honeybee, Apis mellifera. Biocontrol Sci. Technol. 2001, 11, 429–448. [Google Scholar] [CrossRef]
Table 1. Overview of the main results obtained using LABs as probiotics in the honey bee diet.
Table 1. Overview of the main results obtained using LABs as probiotics in the honey bee diet.
LAB SpeciesSourceRelevant Reported ResultsRef.
L. kunkeei
F. fructosus
Honey bee
gastrointestinal tract
Decreases of the mortality rate and significant enhancement of the longevity of honey bees.[64]
L. johnsoniiQueen egg-laying stimulation; higher number of honey bees and a significant increase in honey yield, healthier bee colony[65,66,67]
L. johnsonii
L. kunkeei
L. plantarum
L. salivarius
Increased honey production[42,68]
Bifidobacterium spp.
Lactobacillus spp.
Mild increment in bee survival[69]
B. lactis
L. acidophilus
L. casei
Commercial
probiotic product
Enhancement of bee health. Increased honey production and size of the wax cells[70,71,72]
B. bifidum
L. acidophilus
L. delbrueckii sub. bulgaricus
Administration in pollen substitute resulted in an increase in dry mass and crude fat level[73]
L. brevisIncreased expression of genes encoding antimicrobial peptides (abaecin, defensin-1)[74]
L. plantarum,
L. rhamnosus
Mitigate antibiotic-associated microbiota dysbiosis and immune deficits in adult workers[75]
LAB mix:
B. breve
B. longum
L. acidophilus
L. casei
L. plantarum
L. rhamnosus
Commercial
probiotic product
Enhance honey bee immunity. Higher levels for abaecin and defensin in honey bee larvae[76]
Multiple LAB speciesBo0sting colonies’ strength. Positive physiological changes in probiotic-treated groups of adult bees[77]
B. bifidum
E. faecium
L. acidophilus
P. acidilactici
Advantages of probiotic supplementation include better bee survival and higher dry mass and crude fat level[73]
B. asteroids
F. fructosus
F. pseudoficulneus
F. tropaeoli
Honey bee
hive
Induced immune stimulation (higher level of Apidaecin1). Results suggest that the bee immune response to endogenous bacteria is species-specific[78]
L. kunkeeiMitigate antibiotic-associated microbiota dysbiosis[75]
Fructobacillus spp.Able to utilize lignin and promote the growth of honey bee gut community members[79]
E. thailandicus

L. curvatus
W. cibaria
W. viridescens
W. paramesenteroides
Different
sources
The transcription levels of antimicrobial peptide genes, such as abaecin, defensin, and hymenoptaecin, were found to increase significantly[80]
Taxonomic references: Lactobacillus kunkeei: Apilactobacillus kunkeei; Lactobacillus plantarum: Lactiplantibacillus plantarum; Lactobacillus salivarius: Ligilactobacillus salivarius.
Table 2. Overview of the main results obtained using LABs for the control of the honey bee diseases.
Table 2. Overview of the main results obtained using LABs for the control of the honey bee diseases.
DiseaseLAB SpeciesSourceRelevant Reported ResultsRef.
ChalkbroodAl. kunkeei
L. plantarum
Honey bee
gastrointestinal tract
In vitro growth inhibition of A. apis[154,176,177,178]
L. helsingborgensis
Al. kunkeei
L. melliventris
Reduced in situ larval mummification by percentages greater than 80%.[176,179]
American Foulbrood (AFB)Al. kunkeei
L. brevis
L. plantarum
L. amylovorus
L. fructivorans
L. gasseri
L. kunkeei
L. apis
Honey bee
gastrointestinal tract
In vitro growth inhibition of P. larvae[177,180,181,182]
B. asteroids
B. coryneforme
L. kunkeei
and six Lactobacillus spp.
Reduced honey bee larvae mortality.[183,184]
American Foulbrood (AFB)B. asteroides
B. coryneforme
L. apinorum
L. apis
L. kunkeei
L. kimbladii
L. kullabergensis
L. helsingborgensis
L.mellis
L.mellifer
L.melliventris
Honey bee
gastrointestinal tract
The secretome of the LAB mixture strongly inhibited P. larvae vegetative growth.[185]
L. kunkeei
L. plantarum
L. rhamnosus
Honey bee
hive
Reduced pathogen load and improved survival during P. larvae infection[186,187]
Enterococcus spp.In vitro growth inhibition of P. larvae due to the production of bacteriocins.[188]
E. durans[189,190]
E. thailandicus
L. curvatus
L. plantarum
W. cibaria
W. paramesenteroides
W. viridescens
Different sourcesIn vitro growth inhibition of P. larvae[80,155]
Enterococcus spp.In vitro growth inhibition of P. larvae due to bacteriocin production[191]
European foulbrood (EFB)L. apisHoney bee
gastrointestinal tract
In vitro growth inhibition of M. plutonius.[182]
L. kunkeei and
other thirteen LABs
Administration of LAB supplemented food in vivo and in vitro caused partial and total growth inhibition, respectively, and following decreases in the mortality rate[192]
Al. kunkeeiPurified bacteriocin (kunkecin A) exhibited high antibacterial activity against M. plutonius[193]
L. kunkeeiHoneyAntibacterial activity against M. plutonius[46]
NosemosisL. johnsoniiHoney bee
gastrointestinal tract
Reduction in Nosema spp. spores in hives[66]
L. kunkeeiDecreased the count of N. ceranae spores from adult honey bees.[187]
L. salivariusSignificant decrease in the spore levels of Nosema spp.[194]
L. johnsoniiIn syrup administration of the metabolites produced by L. johnsonii (mainly organic acids), reduced the intensity of the disease[67]
B. asteroides
B. coryneforme
B. indicum
L. johnsonii
L. kunkeei
L. plantarum
Honey bee
hive
Reduction in N. ceranae spores[195]
Enterococcus spp.Honey
bee bread
The CFS (cell-free supernatant) does not affect spore viability as well as N. ceranae development[196]
L. casei
L. plantarum
Commercial
probiotic products
Higher mortality in honey bees treated with the probiotic formula, caused by an increase in the Nosema spp. infection[197]
E. faeciumReduced N. ceranae proliferation and infected honey bees increased survivorship[198]
L. rhamnosusLower survival of honey bees fed with probiotic; rapid development of nosemosis in bees fed with the probiotic[199]
P. acidilacticiIn vivo experimental infection by N. ceranae, showing a significant increase in survival rate (20–30%)[200]
Multiple
species of LAB
Reduction in spore counts in colonies[77]
P. acidilactici
L. plantarum
Regulate genes involved in honey bee development (vitellogenin), immunity (serine protease 40, defensin), and possibly prevent infection by the parasite N. ceranae[131]
VarroosisL. salivariusHoney bee
gastrointestinal tract
In situ reduction in the levels of varroosis between 50 and 80%[194]
E. faecium
L. johnsonii
Enhance honey bee survival and increase their soluble proteins[201]
B. asteroids
L. kunkeei
Surface of freshly collected beesCaused 95–100% mortality of mites in 3 days[202]
Taxonomic references: Lactobacillus kunkeei: Apilactobacillus kunkeei; Lactobacillus plantarum: Lactiplantibacillus plantarum; Lactobacillus salivarius: Ligilactobacillus salivarius.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Iorizzo, M.; Letizia, F.; Ganassi, S.; Testa, B.; Petrarca, S.; Albanese, G.; Di Criscio, D.; De Cristofaro, A. Functional Properties and Antimicrobial Activity from Lactic Acid Bacteria as Resources to Improve the Health and Welfare of Honey Bees. Insects 2022, 13, 308. https://doi.org/10.3390/insects13030308

AMA Style

Iorizzo M, Letizia F, Ganassi S, Testa B, Petrarca S, Albanese G, Di Criscio D, De Cristofaro A. Functional Properties and Antimicrobial Activity from Lactic Acid Bacteria as Resources to Improve the Health and Welfare of Honey Bees. Insects. 2022; 13(3):308. https://doi.org/10.3390/insects13030308

Chicago/Turabian Style

Iorizzo, Massimo, Francesco Letizia, Sonia Ganassi, Bruno Testa, Sonia Petrarca, Gianluca Albanese, Dalila Di Criscio, and Antonio De Cristofaro. 2022. "Functional Properties and Antimicrobial Activity from Lactic Acid Bacteria as Resources to Improve the Health and Welfare of Honey Bees" Insects 13, no. 3: 308. https://doi.org/10.3390/insects13030308

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