Next Article in Journal
Measuring the Rise and Fall of Plasma 25-Hydroxyvitamin D Concentrations in Blue-Tongued Skinks (Tiliqua scincoides) Following Ultraviolet B Exposure and Withdrawal
Previous Article in Journal
Impact of Two Surgical Techniques for Umbilical Hernia Repair, With and Without Peritoneal Opening, on Pain Response, Sedation, and Oxidative Stress in Calves
Previous Article in Special Issue
Human–Chelonian Bond in Italy: An Exploratory Study of Pet Turtle and Tortoise Ownership
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutraceuticals, Social Interaction, and Psychophysiological Influence on Pet Health and Well-Being: Focus on Dogs and Cats

by
Mario Nicotra
1,
Tommaso Iannitti
2 and
Alessandro Di Cerbo
1,*
1
School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
2
Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
*
Author to whom correspondence should be addressed.
Vet. Sci. 2025, 12(10), 964; https://doi.org/10.3390/vetsci12100964
Submission received: 21 August 2025 / Revised: 28 September 2025 / Accepted: 29 September 2025 / Published: 9 October 2025

Abstract

Simple Summary

Pet humanization has transformed animal healthcare and highlighted the importance of nutrition in promoting human–pet social interaction, animal psychophysical well-being and, possibly, longevity. This review examines the impact of nutraceuticals, such as omega-3 fatty acids, pre- and probiotics, plant extracts and dietary supplements in enhancing the human–animal bond by reducing stress and modulating the gut–brain axis, managing bone, skin, and immune diseases and even gastrointestinal disturbs.

Abstract

Pet humanization, particularly in dogs and cats, has transformed animal healthcare and highlighted the importance of nutrition in promoting human–pet social interaction, pet psychophysical well-being and, possibly, longevity. Nutraceuticals, such as omega-3 fatty acids, prebiotics, probiotics, plant extracts and dietary supplements, are endowed with antioxidant, anti-inflammatory, immune-modulating, cognitive-enhancing and gut-microbiota balancing properties. These effects have been shown to contribute to the possible prevention and management of bone and skin diseases, as well as gastrointestinal and behavioral disturbs. Moreover, the human–animal bond has been shown to play a pivotal role in reducing stress, improving sociability, and modulating pets’ emotional and physiological states. Evidence also suggests that nutrition and social interactions can influence the gut–brain axis, impacting the behavior, cognition, and resilience to stress-related disorders. Besides underlining the value of nutraceutical integration into pet nutrition strategies and offering a comprehensive, evidence-based perspective on their potential in improving animal welfare, literature reports about drawbacks of the use/misuse of such substances have been reported.

1. Introduction

The global pet population is estimated to be approximately 703.3 million individuals [1,2]. A recent key factor contributing to the rise in the pet population was the COVID-19 global pandemic, as interest in pet adoption, primarily of dogs and cats, increased during lockdowns [3].
This growth reflects a substantial shift in the human–animal bond. Indeed, whereas domestic animals were once mainly seen as mere working animals, such as watchdogs or mousers, they are now perceived as integral family members and therefore humanized [4,5,6]. Consequently, pet owners are increasingly concerned about their pets’ health and dietary habits [7], paying more attention to the ingredient list and manufacturing process [8], continuously seeking out the best food products that combine high quality, safety, and adequate nutritional value [9,10].
The “anthropomorphization” of pets [11] also contributed to the spread of alternative dietary regimes, including homemade as well as plant- and raw meat-based diets [12], probably due to two main factors: a growing wariness against commercial pet food and the pet food industry, and the enjoyment from preparing food for their pets [13]. In addition, a growing interest in natural ingredients has also been registered, as the inclusion of natural products is widely regarded as a reliable indicator of quality [14].
In this sense, nutraceuticals have gained increasing attention in human [15] nutrition for their safety, nutritional profile, and potential therapeutic applications. This neologism refers to a group of compounds that confer health benefits and play a role in the prevention, management, or treatment of illness [16,17,18,19].
Nutraceuticals are from natural origin (mostly plants, algae, or fungi) and include vitamins [20,21,22], minerals [23], healthy fatty acids [24], polyphenols [25,26,27], glucosinolates [28], terpenoids [29], and alkaloids [16,30]. They have become popular in human nutrition due to their positive influence in several pathological contexts such as diabetes [31], cancer [32,33,34], atherosclerosis [35,36], as well as cardiovascular [17,37] and neurological [18,38] disorders.
Similarly, over the past three decades, their popularity has also increased in veterinary medicine [39], particularly in dogs, cats, and horses [40,41], with successful results on skin [42,43] and ocular disorders [44,45], auricular issues [46], behavioral [47,48] and reproductive [49,50] disturbances, as well as chronic pathologies [51,52,53,54,55]. The great potential and importance of nutraceuticals have also been perceived by pet food industry, which has started incorporating them into pet food formulations. This has created a market with an estimated value of nearly $6 billion in 2023, projected to exceed $10.5 billion by 2035 [56,57]. Concurrently, veterinarians have progressively understood their worth, too, and often employ them, alone or in conjunction with pharmaceutical drugs, to guarantee their clients’ and their own animals’ well-being [58].
This review aims to highlight the main nutraceutical compounds in veterinary medicine and their benefits for companion animals’ health as well as their potential in possibly improving the social interactions.

1.1. Diet and Social Interactions

The social perception of animals has changed over time. In contemporary society, companion animals, particularly dogs and cats, are recognized as integral members of the family [5,6]. Their owners establish a profound emotional bond with them, whose intensity is comparable to that shared with human family members [59].
This connection exhibits a dualistic nature, holding both positive and negative aspects for humans and animals. On the one hand, pets receive food, water, and protection from pain, and diseases [5], while owners, in turn, receive love, physical and psychological benefits [59,60,61]. This aspect led to the creation of the term “Zooeyia” [62], which refers to all the health benefits humans gain from interacting with their companion animals. On the other hand, sometimes the owners run the risk of overhumanizing their pets, which can lead to serious issues, such as wrong feeding habits and onset of obesity and diabetes, and welfare impairment, dressing pets or failing to give them enough physical activity [61].
In this sense, food can play a crucial role in the pet-owner relationship, not only as a means to show love and affection but also as a source to compensate potential nutritional deficits [63,64,65]. Pets supplemented with a high-quality diet can positively improve their sociability, making them more prone to human contact and reducing behavioral issues, especially in dogs [66]. Food can also strengthen the bond between pets and their owners [67] or serve as a positive reinforcement to reward desirable behaviors [68].
Furthermore, it can be strategically employed to mitigate behaviors that disrupt cohabitation between owners and their pets, such as cats’ nocturnal activity [68], thereby improving the overall quality of shared living. In this context, nutraceuticals hold great potential in managing specific pet conditions, e.g., halitosis, that pose a significant problem in the social interaction with their owners [53]. For instance, the supplementation with a nutraceutical diet including Ribes nigrum, Salvia officinalis, Thymus vulgaris, and egg albumen effectively relieved halitosis in dogs [53,54]. Another factor that could seriously impact pet-owner relationship is stress [69,70], a complex response of the organism following exposure to a hostile environment or noxious physical or psychological stimuli [71,72]. It is commonly associated with behavioral expressions, including auto-grooming, body shaking, defecating, urinating, circling, floor licking, and vocalizing, that may alter the daily social equilibrium in the long run [73,74].
In this regards, chewable tablets with Rhodiola rosea and Passiflora incarnata, niacinamide, phospholipids, L-tryptophan, and thiamine hydrochloride, significantly improved stress-related responses in dogs with a behavioral history of stress by significantly reducing the mean stress severity score per stressor and noise sensitivity [70]. It can be therefore argued that food can contribute to the strengthening the pet–owner relationship but also help in overcoming some annoying behaviors that may threaten the solidity and durability of this highly complex bond.

1.2. Diet and Longevity

Improving health and while, possibly, extending longevity have become of utmost importance for scientists in both humans and pets [75,76,77]. Adopting a healthy lifestyle, including balanced dietary habits, is crucial to prevent the onset of chronic diseases and extend lifespan, ensuring a “healthy longevity” [78,79,80].
In recent decades, there has been a notable increase in the life expectancy of companion animals, thanks to advancements in veterinary medicine and nutrition, as well as the adoption of more responsible ownership practices [81]. Adequate nutrition, along with supplementation of nutraceuticals, have been associated with reduced onset of diseases as well as improved sociability and quality of life, thus possibly contributing to longevity of pets [76,77,82,83,84].
Dietary interventions in cats and dogs have been shown to delay the aging process and prevent specific age-related metabolic alterations, particularly those affecting the renal, pulmonary, cardiovascular, and gastrointestinal systems, thereby improving their health and extending their lifespan [81,85,86,87,88].
Among the various dietary strategies employed to enhance lifespan, caloric restriction (CR), for instance, has been linked to improved health outcomes and increased longevity [89,90] through multiple mechanisms, including a reduction in oxidative damage [91], a decrease in body temperature [92], and modulation of sirtuin activity [93]. In this regard, polyphenols, alkaloids, carotenoids, and hormones have been shown to modulate the same cellular and physiological pathways involved in CR, thereby mimicking its beneficial effects and potentially promoting longevity [94].
Also, oils, extracts, or other derivatives from different plants, such as flaxseed, sunflowers, Black Ginseng, Yucca schidigera, chestnut, curcumin, and fructans, significantly improved the well-being, and the health of gastrointestinal, immune, and cardiovascular systems [94,95,96,97,98,99,100,101,102,103].
Other nutraceuticals, such as resveratrol, hawthorn leaves flavonoids, grape seed proanthocyanidin, tea polyphenols, and Rhus verniciflua, resulted effective in counteracting pathological conditions such as poisonings, metabolic, vascular, and digestive issues [104,105,106,107,108]. Consequently, all these favorable outcomes positively improved pet’s behavior and, in turn, their relationship with owners.

1.3. Human–Animal Bonding and Pets’ Well-Being

The first studies concerning the impact that interacting with humans has on animals date back to the late ‘60s, and were conducted on experimental animals [109,110]. In 1966, Lynch and McCarthy exposed nine dogs to human presence and a stressor stimulus (i.e., electrical stimulation) and observed a decrease in heart frequency and the suppression of the foot flexion reflex if the animal was petted for 10 s while receiving the stressor [110]. Both signs were associated with a positive impact on human-pet interaction, possibly indicating an animal more prone to establish a relationship. Being more than an isolated case, the study from Lynch and McCarthy fostered similar future studies, which markedly raised the interest in societal perception of pets in recent decades, thus emphasizing the respective positive impact of human–animal interaction. Simple acts, such as petting, training, or playing with them, can help pets alleviate their stress [111], possibly modulating also some behavioral disorders including anxiety, fear, and hyperactivity [48]. The stress-attenuating effect due to the interaction was also evident while assessing the impact of verbal and tactile interaction between owners and their pets during a veterinary clinical examination [112]. Results revealed a reduction in examination-related stress, as indicated by fewer attempts to jump off the exam table, a lower heart rate, and a lower maximum ocular surface temperature in dogs receiving tactile and verbal support from their owners.
A similar positive impact on stress was observed in sheltered animals [113,114]. Two in vivo studies found that human interaction could reduce stress in these animals, as confirmed by improvements in sociability, diffidence, temperament, excitation, and vocalization, and panting, followed by reductions in cortisol levels in blood plasma and saliva [113,114].
Human-pet bond can also influence the blood concentration of oxytocin, a pivotal hormone involved in the establishment and duration of the relationship [115], whose increased and rapid release is a consequence of positive emotions such as affection (i.e., kisses), love and physical interaction [116,117,118].
Similarly to dogs, the interaction with humans has also been linked to reduced stress and anxiety in cats. It has been proposed as an alternative strategy to mitigate stressful behaviors, and research has shown that gentling can also enhance the production of S-IgA and prevent the onset of upper respiratory diseases [119,120,121]. All this evidence supports the importance that the human–animal bond holds not only for human health but also for the well-being of companion animals. Thus, emotional factors such as love and care can positively affect the well-being of humans and pets by decreasing stress and, if combined with a diet enriched with antioxidants, improving the quality of life and promoting longevity.

2. Materials and Methods

This review was conducted to comprehensively and critically summarize the scientific literature, examining the role of nutraceuticals in enhancing pet longevity and influencing social behaviors and interactions with humans. Relevant peer-reviewed publications were identified through searches of electronic databases, including PubMed, Web of Science, Scopus, and Google Scholar, covering the period from January 2000 through June 2025. Search terms combined key concepts related to companion animals, nutraceuticals, longevity, aging, cognition, and social behavior, including: “nutraceuticals” AND “pet food”, “functional ingredients” AND “dogs AND cats”; “dietary supplements” AND “companion animals” AND “Animal welfare” AND “Emotional bonding” AND “Proper animal care” AND “Social interaction” AND “Physical activity support” AND “Quality of life improvement” AND “Antioxidant protection” AND “Cellular protection” AND “Longevity promotion” AND “Omega-3 fatty acids” AND “Anti-inflammatory support” AND “Balanced phosphorus” AND “Renal (kidney) health” AND “Holistic nutrition” AND “Integrated care” AND “Nutritional balance” AND “Synergistic effects of nutrients” AND “longevity”. Boolean operators (“AND,” “OR”) were used to refine the queries. Additional references were retrieved by cross-referencing citations in relevant articles and reviews.
Based on the results retrieved from the literature, we first summarized the mechanism of action, scientific evidence, species-specific considerations, and commercial availability of the main nutraceutical substances used in the pet food industry, such as omega-3 fatty acids, prebiotics and probiotics, antioxidants, plant-based extracts, and dietary supplements such as vitamins, minerals, glutathione, glucosamine, and chondroitin sulphate.
Then, we described their health benefits in the main pet apparatuses, such as joints, skin and coat, gut, cognitive functions, and immunomodulation related to pet longevity and social interactions.

3. Results and Discussion

3.1. Omega-3 Fatty Acids

Polyunsaturated fatty acids, or “PUFAs”, consist of a long chain of carbon atoms, a carboxyl group on one end, and a methyl group on the other. They exhibit at least two double bonds between the carbons of the chain and are classified as Omega-3 (n-3) and Omega-6 (n-6) fatty acids, respectively, based on the presence of a double bond on the third or sixth carbon atom from the methyl end [122]. N-3 fatty acids can be further distinguished into short-chain (SC), such as alpha-linolenic acid (ALA; 18 carbon atoms), or long-chain (LC), including eicosapentaenoic (EPA; 20 carbon atoms) and docosahexaenoic (DHA; 22 carbon atoms) acids [122,123].
ALA is abundant in plant oils, such as flaxseed, walnuts, soybeans, hemp, rapeseed, chia, canola, and perilla oils [124,125], while fish and seafood are rich in EPA and DHA [126]. In humans as well as in mammalian species, ALA can be converted into EPA and DHA [127]. However, this conversion is not efficient enough to meet the adequate requirements for EPA and DHA, which are considered essential nutrients and must be supplied through the diet [120,121,122,123,124,125,126,127,128,129,130,131].

Health Benefits of Omega-3 Fatty Acids and Mechanisms of Action

Omega-3 fatty acids offer several health benefits for humans and animals, which can help increase their lifespan and promote longevity, while also enhancing their overall quality of life. First, they are endowed with anti-inflammatory and immune-modulating properties [132]. These activities can be related to several mechanisms, including the modulation of inflammatory mediator production, such as eicosanoids, tumor necrosis factor alpha (TNF-α), ad interleukin 6 (IL-6) [133], and the impairment of leukocyte adhesion to endothelium due to the reduced expression of adhesive surface molecules on endothelial cells [134,135], monocytes [136], macrophages, [137] and lymphocytes [138]. In addition, another hypothesized anti-inflammatory mechanism is related to the production of n-3 PUFAs metabolites (resolvins D and E, marensins, and protenectins) by specific enzymes such as lipoxygenases and cytochrome P450 [139].
The anti-inflammatory effects of n-3 PUFAs may also be responsible for the amelioration of clinical conditions and the faster reduction in carprofen need in dogs with osteoarthritis [140,141].
Omega-3 fatty acids also support the health of skin and hair coat, especially in animals suffering from allergies or other inflammatory skin conditions, by competing with arachidonic acid (AA) for cyclo-oxygenase and 5-lypoxygenase and leading to the production of eicosanoids that exert anti-inflammatory activity or have weaker pro-inflammatory activity than the ones derived from AA [142,143,144]. Moreover, high DHA was observed to reduce excessive skin oil secretion, thereby supporting coat quality [145].
They can also enhance cardiovascular functions through various mechanisms. For instance, they have antiarrhythmic properties that are likely due to their modulation of ionic currents, including L-type calcium currents, in cardiac cells [146,147]. They can also reduce blood pressure and protect against ischemic damage by modulating leukocyte function and reducing their infiltration into ischemic tissue [146,148].
In addition, n-3 fatty acids impact reproduction by influencing the production of steroid hormones, modulating the secretion of molecules involved in reproduction, such as eicosanoids like PGE2 and PGFα, and affecting the functionality of sperm and oocytes [149]. In females, they influence the development of oocytes and support the germinal vesicles, thereby preventing them from breaking down [150]. Moreover, they inhibit cyclooxygenase, modulating the production of prostaglandins, including PGE2 and PGFα, which play a key role in the development, maintenance, and regression of the corpus luteum. In males, on the other hand, n-3 fatty acids have been associated with the improvement of several characteristics of spermatozoa, including increased membrane fluidity, motility, morphology, and concentration [151]. On the contrary, dietary restriction of n-3 PUFAs was linked to poor semen quality, and reduced levels of EPA and DHA were found in sperm samples from infertile patients [49,151].
Omega-3 supplementation has also been shown to exert renoprotective effects in dogs by counteracting hypercholesterolemia and hypertriglyceridemia, both of which had progressively negatively impacted renal health [152]. Moreover, these protective effects can also be attributed to the omega-3-induced increase in glomerular filtration rate (GFR), reduction in oxidative damage, and suppression of inflammation [153].
Finally, n-3 PUFAs also exert several benefits on brain development and vision, both in young and aging patients [154]. DHA concentrations increase during perinatal brain development, reaching around 1020% of its total fatty acid composition 21 days after birth. They support the maturation of the brain, enhancing the formation of synapses and the development of neuronal arborization [155]. Furthermore, EPA and DHA have been shown to positively affect cognition in aging dogs and cats, likely due to their protective effects against inflammation and oxidative stress, as well as their ability to enhance neurogenesis and glucose transporter activity [156]. However, omega-3 supplementation is not free from side effects. Indeed, research reports potential adverse effects, including weight gain, impact on insulin sensitivity, alterations in platelet function and wound healing, diarrhea, pancreatitis, and lipid peroxidation, even though studies are conflicting. Moreover, Omega-3 supplementation can increase the risk of falling into fat-soluble vitamins hypervitaminosis, alterations in immune function, with decreased neutrophil function and lymphocyte counts, and a negative impact on hypersensitivity reactions and nutrient–drug interactions, especially when administered concurrently with NSAIDs, such as Carprofen [29].
All this evidence shows the Importance of omega-3 PUFAs in companion animals’ health and their potential to prevent the onset of pathological conditions.

3.2. Prebiotics and Probiotics

The term “probiotics” refers to live microorganisms that positively affect the host’s health if supplemented in proper amounts [157,158]. They primarily act by improving intestinal microbial balance, leading to both intestinal and gastrointestinal benefits, including immune-modulating [157]. The most popular probiotics for dogs and cats come from the Lactobacillus and Bifidobacterium genera (e.g., Bifidobacterium animalis subspecies lactis CECT 8145 [159], Bacillus velezensis DSM 15544 [160]). Recently, however, researchers have begun investigating yeasts and other microorganisms, such as Saccharomyces cerevisiae and Akkermansia muciniphila, Faecalibacterium prausnitzii, Eubacterium hallii, Prevotella copri, Christensella minuta, Parabacteroides goldsteniii, and species belonging to the Bacteroides genus for their probiotic properties [161,162,163,164]. Many of the prebiotics used to ensure the health and well-being of pets are of human origin [165,166]. Nevertheless, the focus of research has shifted to isolating potential probiotic strains from animal hosts, aiming to enhance further their safety and efficacy in promoting health in companion animals [167,168].
Prebiotics, on the other hand, are non-digestible nutrient substrates selectively employed by the microorganisms present in the host’s gut microbiota. Their degradation products, mainly short-chain fatty acids, enter the bloodstream, thereby potentially extending their health benefits to other districts beyond the gastrointestinal tract [168,169,170].
The most common prebiotics employed in animal nutrition include fructans, such as fructooligosaccharides (FOS) and inulin, galactans, including galactooligosaccharides (GOS), Xylooligosaccharides (XOS), lacticol, and cereal fibres [171].
Pre- and probiotics work synergistically; as a result, they are frequently combined in formulations known as “synbiotics” [172,173].

Health Benefits of Pre- and Probiotics and Mechanisms of Action

Ensuring an optimal balance within the gut microbiota is crucial for maintaining the host’s health [174]. Alteration to its homeostasis, known as “dysbiosis”, can significantly affect an animal’s well-being and increase the risk of developing pathological conditions, including inflammatory bowel disease (IBD), obesity, or neurological disorders [175].
Companion animals can benefit positively from pre- and probiotic supplementation. Prebiotic supplementation contributes to intestinal health by promoting the proliferation and activity of beneficial intestinal microbes, such as bifidobacteria and lactobacilli, thus positively impacting gut microbiota. Moreover, they can also be employed to modulate fecal pH, consistency and bulk, and increase short-chain fatty acids (SCFA) production [176]. In dogs, as in humans, gut microbes can ferment prebiotics, thereby producing acetate and lactate that serve as substrates for other microbial species, which utilize them to produce butyrate, another SCFA.
Butyrate represents the primary energy source for colonocytes, contributes to intestinal well-being, and exhibits immunomodulatory properties by modulating the production of IL-2 and IL-6 [177].
Prebiotics can also play a role in the prevention of certain disorders, such as obesity, which could potentially facilitate the onset of chronic pathologies, including diabetes mellitus. Indeed fibers, particularly those with high fermentability, can enhance satiety, thereby reducing daily energy intake and promoting weight management [178]. Moreover, prebiotics can positively impact insulin sensitivity, glucagon-like peptide-1 (GLP-1) secretion, and postprandial hyperglycemia [179,180], thus representing a potential means to prevent the onset of diabetes. In addition, despite their capacity to modulate gut microflora and restore SCFA production, prebiotics are often used, in combination with probiotics, to counteract gastrointestinal diseases and their clinical signs, including diarrhea [181].
Likewise, probiotic supplementation affects gut microbiota and plays a pivotal role in rebalancing the gut microbiota of companion animals with gastrointestinal disorders [161]. Probiotics enhance the host’s health through different mechanisms. For instance, they can interact synergistically with the gut microbiota, supporting its stability and cooperating in the metabolization of nutrients. They can also prevent pathogens from colonizing the gut by stimulating the synthesis of antimicrobial compounds, such as bacteriocins, and by competing for nutrients and sites, thereby exerting an overall protective action [182]. Moreover, they can ameliorate the intestinal barrier integrity by stimulating mucin production, promoting the well-being of intestinal epithelial cells, and reducing the production of pro-inflammatory cytokines [183,184].
In both dogs and cats, probiotics have been shown to improve fecal quality and reduce fecal nitrogen fermentation products, ammonia emissions, fecal inflammatory markers, and overall fecal odor, while increasing fecal antioxidant and the production of SCFAs [185,186,187,188,189]. In addition, their potential therapeutic use has been proposed in the management of several gastrointestinal disorders, including colonic dysmotility [190] and diarrhea [191,192] in dogs. Furthermore, research in felines reports their positive impact on managing oral issues, such as stomatitis, gingivostomatitis, and both bacterial and viral oral infections, as well as improving immunity, hepatic, and renal health [193,194]. For instance, cats with chronic kidney disease (CKD) at stage 2 or 3 supplemented for two months with a Lactobacillus mixture showed reduced or preserved concentrations of the blood urea nitrogen (BUN) and creatinine, the two leading kidney function indicators in blood plasma, and indoxyl sulfate, a gut-related uremic toxin whose increase in circulation is negatively correlated with the progression of CKD. Furthermore, the treatment resulted in increased appetite, activity, and defecation frequencies, thereby contributing to an overall enhancement of the patients’ quality of life [194].
The influence of probiotics on microbiota composition can also indirectly impact animals’ behavior by affecting the bidirectional communication system, which links the intestinal and nervous systems, known as the “gut–brain axis” (GBA) [195].
Indeed, modifications in the intestinal microbial composition have been linked with behavioral issues, such as aggressiveness and anxiety [196,197,198]. In this context, probiotic supplementation has shown beneficial effects on modulating behavioral disorders, thus representing a promising nutraceutical option in the treatment of these conditions [199,200]. For instance, daily supplementation with Lactobacillus plantarum LP815TM for four weeks improved aggressiveness and anxiety in 40 dogs effectively. There were no adverse effects, and sleep regularity was enhanced as an additional benefit [200].
The use of pre- and probiotics is generally considered safe. However, prebiotics misuse can lead to flatulence, diarrhea, and abdominal pain [201], while probiotic supplementation, although occasionally, can result in opportunistic infections, and in immunological and metabolic disturbances [202]. Furthermore, probiotics can carry antimicrobial resistance genes (ARGs), particularly those belonging to the genus Enterococcus, thus contributing to the spread of antimicrobial resistance (AMR) [202,203,204].
In any case, despite the absence of a specific recommendation, research findings indicate that the provision of pre- and probiotics to companion animals exerts a direct influence not only on their gastrointestinal health. Furthermore, it can indirectly engender extraintestinal favorable outcomes, such as those observed in behavior, promoting animal health and ensuring their longevity.

3.3. Plant Extracts

This increased interest in medicinal plants and herbal extracts stems from the perception that natural compounds are safer, more economical, and environmentally friendly than their synthetic counterparts, yet are just as effective [205]. Medicinal plants hold great potential in veterinary medicine because they produce phytocompounds, such as polyphenols (e.g., flavones, flavanones, and flavanols), carotenoids (e.g., lutein and lycopene), isoprenoids (e.g., limonene and pinene), phytosterols (e.g., campesterol and sitosterol), saponins (e.g., dammarane and oleanane), dietary fibers (e.g., pectin and cellulose), and specific polysaccharides (e.g., amylose and amylopectin) that confer several health-enhancing biological properties [206].
These phytochemicals are each associated with specific health-enhancing properties, including antimicrobial and immunomodulatory activities, stress relief, and growth promotion [207]. Polyphenols, for example, are secondary metabolites of plants and are abundant in foods of vegetable origin, such as fruit (e.g., blackberries, strawberries, kiwi, cherry, peach, apricots, apples, etc.), vegetables (e.g., aubergines, artichokes, potatoes, leeks, etc.), cereals, nuts, and legumes [208,209]. They are well known for their antioxidant activity, but they possess other bioactivities, including anti-inflammatory, cardioprotective, antineoplastic, and antimicrobial properties [210].
Carotenoids, such as β-carotene, are one of the primary dietary sources of vitamin A. They are abundant in fruits (e.g., apricots and peaches), green vegetables (e.g., spinach and broccoli), along with some foods of animal origin, including butterfat, egg yolk, and salmon. These compounds have been associated with several health-enhancing properties, including antioxidant, immunomodulatory, cardioprotective, and antineoplastic activities [211].
Phytosterols, on the other hand, are steroids of natural origin endowed with chemopreventive, antioxidant, anti-inflammatory, and antidiabetic effects [212], also able to impact the metabolism of cholesterol in humans and dogs [210,213].

Health Benefits of Plant Extracts and Mechanisms of Action

Plant extracts can be used either to improve the well-being of healthy animals, alone or in combination with other substances, by enhancing immune, gastrointestinal, and cardiovascular health, as well as contrasting pathological conditions such as heavy metal poisonings, metabolic issues, including obesity, and diseases involving the digestive tract [76]. The health benefits of several plant extracts have been investigated for their potential therapeutic use in veterinary medicine [214].
For example, Kim et al. (2020) assessed the anti-inflammatory potential in beagle dogs fed two different doses of black ginseng extract for 8 weeks by evaluating their serum metabolic profile [97]. Results have shown a statistically significant difference in the metabolic profiles of treated and untreated dogs, which is hypothesized to be linked to its biological activity. The anti-inflammatory effect is probably due to the presence of saponins, such as ginsenoside Rh2, Rg1, Rb1 and Rg3, that inhibit the production of some pro-inflammatory mediators, such as nitric oxide (NO), COX-2, and pro-inflammatory cytokines such as TNF-α and interleukin-1β (IL-1β), along with an increase in anti-inflammatory cytokines, specifically IL-10 [215,216].
Likewise, Reichling et al. (2004) investigated the effect of a 42-day administration of a resin extract of Boswellia serrata in dogs with osteoarthritis and spinal disease, demonstrating the potential of this plant extract to improve the clinical condition of dogs with osteoarthritis [214]. The anti-rheumatic and anti-inflammatory properties of this plant-based supplement stem from the presence of several compounds that interfere with the production or action of mediators involved in the inflammatory process, including TNF-α, IL-1β, NO, mitogen-activated protein kinases (MAPKs), gamma-interferon (IFN-γ), and IL-6 [217,218]. Boswellia extract was also associated with increased antioxidant defense and inhibition of lipid peroxidation, thus showing it possesses an overall antioxidant power [218]. However, although being natural, plant extracts are not completely harmless. For instance, the administration of a garlic extract to healthy adult mixed-breed dogs resulted in significantly decreased erythrocyte count, hematocrit, and hemoglobin concentration, along with Heinz bodies formation and increased erythrocytic methemoglobin concentration. Furthermore, morphological abnormalities, including the formation of eccentrocytes, was observed [219]. Moreover, side effects following the supplementation of plant extracts may also derive from the contamination of the material used during the extraction, including also the potential presence of heavy metals, fungi or chemical substances, such as pesticides [220].

3.4. Dietary Supplements

Dietary supplements are substances intended to be administered orally, either with food or separately, and that, despite being different from drugs, produce specific health benefits for the animals to which they are administered [40]. Multivitamins, chondroprotective agents, and glucosamine are among the most popular supplements among pet owners. However, minerals, amino acids, enzymes, organ tissues, and metabolites are also quite common [40,221]. In companion animals, they are often used to support joint health and gastrointestinal function, while also promoting cognitive function, skin and coat health, and the well-being of the cardiovascular and urinary systems [40].

3.4.1. Health Benefits of Vitamins and Mechanisms of Action

Vitamins are small, organic, essential molecules that fulfill important roles in animal organisms at all stages of life. Based on their solubility, they are classified into fat-soluble (A, D, E, K) and water-soluble (B complex and C) vitamins [222,223]. Furthermore, the B complex group includes nine different vitamins, specifically thiamine (B1), Riboflavin (B2), Niacin (B3), Pantothenic acid (B5), Pyridoxine (B6), Biotin (B7), Folate (B9), and Cobalamin (B12) [223]. All of them play essential roles in the health and well-being of animals, thereby supporting their longevity, and their deficiency can lead to serious detrimental consequences.
For instance, vitamin A is involved in the morphogenesis of eyes, the retention of their structural integrity, and in ensuring the correct structure and function of retinal photoreceptors, thereby protecting vision. Furthermore, it has antioxidant and immunomodulating properties, plays a crucial role in cell growth and differentiation, which is particularly relevant during embryo development, and ensures the health of the reproductive tract [224].
Vitamins belonging to the B complex act as cofactors or coenzymes and are involved in the metabolism of carbohydrates, proteins, and lipids [225,226,227].
Vitamin C is known for its antioxidant properties, but it is also involved in tissue growth and maintenance, the modulation of the immune system, and the synthesis of vasoactive substances, including catecholamines and vasopressin [228].
Vitamin D is essential for calcium homeostasis and to ensure the correct development and maintenance of bones [229]. However, hypovitaminosis D has also been observed in extra-skeletal pathologic conditions, including gastrointestinal, renal, cardiac, infectious diseases, cancer, and inflammation. Therefore, research is now focusing on understanding whether vitamin D is somehow linked to the onset of these pathologies or if it can be used as a marker of illness [230].
Vitamin E, also known as alpha-tocopherol, has a potent free radical scavenging activity, thus protecting against oxidative stress. It also works synergistically along with other antioxidant molecules. For instance, vitamin C has been observed to support the regeneration of vitamin E during the process of free radical scavenging, and supplementation with a blend of vitamins B, C, and β-carotene has shown increased cell protection and reduced oxidative damage [231].
Finally, vitamin K is well known for its role in the coagulation cascade and is used in veterinary medicine to treat anticoagulant rodenticide toxicosis in both dogs and cats [232,233]. However, it also has antioxidant and anti-inflammatory activities, as well as protective effects on several districts, including the nervous, cardiovascular, and immune systems, as well as bones, and can reduce the incidence of certain pathological conditions, such as type 2 diabetes mellitus and pathogenic thrombosis [234].
Vitamins play a pivotal role in all animal organisms. However, their metabolism can differ among different species. For instance, dogs and cats are unable to synthesize vitamin B1 [225]. Moreover, compared to other species, they have physiological enzymatic deficiencies that limit their ability to synthesize vitamin D3 [230,235], and cats are unable to convert dietary β-carotene into vitamin A and to synthesize niacin from tryptophan [235].
Hence, vitamin supplementation becomes relevant primarily in cases where species-specific enzymatic limitations pair with suboptimal environmental or nutritional management, that might harm pets’ health and reduce life expectancy by impairing organ function and increasing oxidative and inflammatory damage. Nevertheless, vitamins should be supplemented cautiously, since excessive vitamin intake, especially concerning fat-soluble vitamins, can result in toxicosis, also known as “hypervitaminosis” [236]. Concerning hypervitaminosis A, dogs and cats appear to be more tolerant compared to other domesticated animals; however, they can develop vitamin A toxicosis, with symptoms including diarrhea, reduced appetite, neurological symptoms, bone demineralization, and reduction in thyroxin plasma concentration [224]. Hypervitaminosis D often follows excessive vitamin D dietary supplementation, rodenticide poisoning, or treatment with substances containing vitamin D2, D3, or vitamin D metabolites or analogues. It is one of the main causes of hypercalcemia both in dogs and cats, and symptoms include lethargy, inappetence, gastrointestinal disturbances, such as vomiting and melena, polyuria, and polydipsia [237,238]. Finally, while no hypervitaminosis K has been described, Vitamin E toxicosis is rare in companion animals; however, overdosing can result in bleeding problems due to vitamin K inhibition and impairment of leukocyte and lymphocyte functions [236].

3.4.2. Health Benefits of Minerals and Mechanisms of Action

Minerals are essential micronutrients involved in several biological processes, ranging from initiating hormone production to tissue and subcellular functions [239].
According to the required daily amount, they are classified into two main groups. The “macro elements” include calcium, chloride, magnesium, phosphorus, potassium, and sodium, while the “trace minerals” include iron, zinc, selenium, copper, manganese, and iodine [4,239,240]. Mineral supplementation is essential for maintaining essential bodily processes. However, both excess and deficiency can result in serious health consequences, such as metabolic and neurological disorders, as well as musculoskeletal, cardiovascular, and oncological diseases, in both humans and animals [241,242].
For instance, zinc, copper, and manganese impact the antioxidant system by influencing the activity of superoxide dismutase (SOD) [243]. Zinc is involved in the proper functioning of alkaline phosphatases, and its optimal supplementation has been associated with increased longevity in dogs [244].
Moreover, mineral imbalance has been linked to the onset of several pathologies. For example, zinc deficiency has been associated with skin [245,246,247] and behavioral [248] disorders. Conversely, a high zinc serum concentration results in zinc toxicosis, characterized by inappetence, vomiting, and regenerative anemia [249]. Moreover, research conducted by Vitale et al. (2019) hypothesized that an imbalance of manganese, selenium, and zinc serum concentrations may play a role in canine idiopathic epilepsy [250].
Calcium and phosphorus play a crucial role in the health of dogs and cats. Beyond its involvement in bone development and skeletal health, calcium participates in a variety of processes, including blood coagulation, nerve impulse transmission, cardiac contractility, hormone and neurotransmitter secretion, enzyme reactions, and intestinal motility [251,252]. Phosphorus, on the other hand, contributes to the development of bones and teeth, the maintenance of osmotic and acid-base balance, electrolyte transport, and several enzymatic systems, as well as taking part in the synthesis of nucleic acids [253].
Imbalance in the concentrations of these elements, whether characterized by excess or deficiency, can lead to potentially life-threatening complications requiring emergency treatment and prolonged hospitalization [254,255,256,257]. Moreover, imbalanced levels of calcium and phosphorus, along with dysregulation of vitamin D and parathyroid hormone, can facilitate the onset of chronic kidney disease—mineral and bone disease (CKD-MBD) [258]. Also, selenium is fundamental in keeping humans and animals healthy. It is involved in several mechanisms, including redox balance, immune modulation, DNA synthesis, and ensuring the proper functioning of the reproductive system and thyroid metabolism [259,260,261]. Furthermore, research conducted on canine neoplastic cell cultures has enabled researchers to speculate on the potential role of selenium in cancer prevention [262,263,264].
The aforementioned examples highlight the pivotal role of minerals in maintaining the well-being of companion animals. Therefore, it is evident that ensuring proper mineral supplementation for pets can significantly impact their physical well-being and prevent the onset of potentially life-threatening conditions that could compromise their health and longevity. However, minerals should be supplemented carefully since an excessive intake can lead to severe toxicosis [265], resulting in a wide range of manifestations, including gastrointestinal symptoms, anemia, lethargy, anorexia, and emaciation [261,266,267].

3.4.3. Health Benefits of Glutathione and Mechanisms of Action

Glutathione, also known as gamma-glutamyl-cysteinyl-glycine (GSH), is a tripeptide synthesized primarily in the liver from glutamate, cysteine, and glycine, and is widely recognized for its antioxidant properties [268]. It is synthesized in the cytosol and distributed to intracellular organelles, including mitochondria, which are the primary source of reactive oxygen species (ROS) [269,270]. Besides being the primary intracellular antioxidant agent, it is involved in several cellular functions, including the modulation of cellular proliferation, apoptosis, and gene transcription [271]. Moreover, it also plays a role in the detoxification of xenobiotics and endogenous compounds [269,272]. It exists in two primary forms, reduced (GSH) and oxidized (GSSG) glutathione, whose ratio is used to determine the redox status of the cells. The optimal GSH/GSSG ratio is >100, while a ratio of 1:10 is characteristic of cells exposed to oxidative stress (OS) [269].
GSH exerts its protective effect against cellular oxidative damage both directly and as a cofactor of antioxidant and detoxification enzymes, including glutathione peroxidases, glutathione S-transferases, and glyoxalases [273]. The direct antioxidant and scavenging abilities of GSH are attributable to its capacity to donate electrons, which enables GSH to neutralize ROS, including superoxide, hydroperoxyl, and hydroxyl radicals [274]. Hence, GSH is fundamental to preserving intracellular redox balance and mitigating OS [275].
Obstructing the OS is of utmost importance to ensure pets’ health and longevity, as it has been linked to the onset of a multitude of pathologies in both dogs and cats, such as gastrointestinal, hepatic, pancreatic, and endocrine diseases [259,276,277], and has been recognized as a contributor to aging [278]. All these conditions can influence the quality of life of companion animals and reduce their lifespan. Moreover, reduced GSH concentration has been detected in ill dogs and cats, which was also correlated with illness severity and increased mortality [279]. Particularly, low GSH values were found in both cats and dogs suffering from liver diseases [280]. This can be linked to the fact that the liver is the primary GSH producer [281] and contains the highest GSH concentration; hence, liver dysfunction can cause a reduction in GSH concentration, both due to reduced synthesis and increased use [282]. In human medicine, GSH supplementation is used for its beneficial effects on health, including anti-aging, skin-protective, immune-modulating, and hepatoprotective properties [283,284,285]. In veterinary medicine, glutathione-based supplements are less popular. However, some studies support its usefulness in supporting animal health. For instance, Vulcano et al. (2012) observed that GSH supplementation reduced acetaminophen (APAP)-induced methemoglobin formation and hepatotoxicity in cats, suggesting that GSH can be used as a therapeutic agent to mitigate APAP toxicity [286]. Glutathione supplementation can also be beneficial in dogs by increasing erythrocyte GSH levels and exerting hepatoprotective effects, resulting in improved liver parameters, notably alanine transaminase (ALT), alanine aminotransferase (AST), alkaline phosphatase (ALP), gamma-glutamyl transferase (GGT), and bilirubin (BIL) [287].
Besides being the most important antioxidant molecule of the organism, GSH also supports cellular functions and protects endogenous and exogenous toxic compounds, thereby contributing to pets’ health and longevity.

3.4.4. Health Benefits of Glucosamine and Chondroitin Sulphate and Mechanisms of Action

Glucosamine and chondroitin sulphate (CS) are two nutraceutical substances primarily employed in veterinary medicine for the management of osteoarthritis (OA) due to their chondroprotective properties [288]. CS is a natural glycosaminoglycan that is detectable in all connective tissues, particularly in articular cartilage [289]. Its primary function is to ensure joint health; however, it also possesses antioxidant, anti-inflammatory, and immune-modulating properties [290]. Glucosamine, on the other hand, is an amino sugar that is abundant in articular cartilage, intervertebral discs, and synovial fluid. It can be isolated from the shell of shellfish [291] and is primarily employed orally in the management of OA [292].
CS and glucosamine act synergistically to promote the synthesis of glycosaminoglycan in chondrocytes. Moreover, CS was also associated with the inhibition of collagenase and aggrecanase, two proteolytic enzymes involved in the degenerative processes of cartilage. Overall, the combination of CS and glucosamine hinders cartilage degeneration, thereby positively impacting the progression of OA [293].
OA is a multifactorial joint pathology characterized by articular chronic inflammation and cartilage damage, including irregularity and focal erosions. These alterations gradually spread to the entire joint, resulting in pain and loss of function [294,295], which negatively impacts the quality of life of the affected animals.
OA represents the most commonly diagnosed joint pathology in veterinary medicine. Indeed, it is frequently observed in aging dogs, particularly in large and giant breeds, as well as in cats [295,296,297]. Moreover, OA is recognized as the leading cause of chronic pain [298], which can result in premature euthanasia of companion animals if under-recognized or under-managed [299], thus negatively impacting animals’ longevity.
To date, the use of both CS and glucosamine in the management of OA is controversial, as research conducted has shown heterogeneous results [288,292,300]. Nevertheless, they can mitigate OA damage, thus improving pets’ quality of life. In a recent in vitro study, Bai et al. (2024) observed the ability of CS to suppress the secretion of pro-inflammatory cytokines in lipopolysaccharide (LPS)-challenged canine and feline chondrocytes [301]. Moreover, it also promoted cell viability and proliferation, confirming its ability to modulate inflammation and its potential role in the treatment of OA. Furthermore, in vivo research has highlighted the efficacy of combining glucosamine and chondroitin sulfate in reducing joint pain and their chondroprotective effects in rat models [302,303]. Accordingly, oral administration of a combination of CS and glucosamine in dogs with OA resulted in clinical amelioration and improvements in pain scores [304]. However, results are conflicting, and more research is needed to assess the fundamental role of CS and glucosamine in the management of OA in companion animals [305,306,307,308,309].
Altogether, the vast array of nutraceutical substances, from omega-3 fatty acids to pre- and probiotics, plant extracts, vitamins, minerals, and chondroprotective agents, suggests their pivotal role in promoting systemic health, preventing disease, and ultimately supporting the longevity and quality of life of companion animals.

3.5. GBA and Gut Health Modulation by Nutraceuticals

Increasing evidence suggests that gut microbiota balance and stability, plays a crucial role in maintaining the host’s health and aging-related changes, thus contributing to the human and animal longevity [310,311,312]. Conversely, dysbiosis can lead to the onset of pathological conditions, including mental disorders and anxiety, that negatively influence the pet-owner relationship, potentially leading to abandonment or euthanasia [313].
The gut exerts a dynamic interaction with other organs (liver [314], kidneys [315], lungs [316], and brain [317,318]) through several pathways, referred to as “axes”, positively or negatively modulating their function [319]. Particularly, the GBA is a complex, bidirectional communication system between gut microbiome and the central nervous system (CNS) [320,321], whose interplay is so intimate that the gut is often referred to as a “second brain” [322].
The gut microbiome produces multiple metabolites, neuroactive substances and hormones that can reach the brain through the enteric nervous system (ENS) [323], whose mediator is GABA, the vagus nerve, circulation, or the immune system and modulate its function [324]. On the other hand, the brain can communicate with the gut microbiota by inducing neurons, immune cells, and enterochromaffin cells to secrete signaling molecules, thereby modulating gut functionality and environment (pH, motility, and mucus), and creating optimal conditions for the microflora [325].
The gut microbiota can also influence the production of neurotransmitters such as dopamine (DA), serotonin (5-HT), glutamate, and gamma-aminobutyric acid (GABA), which are involved in brain function, cognition, and social behavior [326,327,328].
Serotonin, for instance, was shown to play a role in circadian rhythms and multiple psychological processes, including mood, perception, reward, anger, aggression, fear, memory, sexuality, addiction, and attention [329]. Conversely, dopamine can regulate various functions, including cognition, emotion, positive reinforcement/reward-driven learning, food intake, and motivation [330,331].
The GBA is a relatively novel concept that has garnered the interest of researchers due to its health-enhancing potential, including a possible role in maintaining homeostasis and modulating behavior [332], anxiety, depression, affect, motivation, and cognitive functions [333]. Particularly, dysregulation of GBA crosstalk has been associated with many pathological conditions, including metabolic syndrome, psychiatric disorders such as depression and anxiety, and autism spectrum disorders (ASD). GBA crosstalk has also been linked to neurodegenerative diseases, such as Parkinson’s disease (PD) and Alzheimer’s disease (AD) [334]. In addition, research has shown that the gut microbiota of people suffering from mental disorders, including anxiety, is altered compared to healthy controls [335,336,337,338].
Similarly to humans, the existence of the gut–brain axis and the link between gut microbiota alterations and pathological conditions has been documented in companion animals [313]. Likewise, in pets, dysbiosis has been linked not only to neurological diseases, such as idiopathic epilepsy [339,340], but also to gastrointestinal [341,342] and metabolic diseases [341], exocrine pancreatic insufficiency [343], behavioral disorders [196,197,198], and cancer [344].
To ensure GBA health, thereby reducing aging-related changes and possibly promoting longevity, a correct nutrition based on nutraceutical substances and specific ingredients that can positively influence the composition of the gut microbiota is mandatory [345].
Nutraceutical substances, including “biotics” (pre-, pro-, post-, and synbiotics), as well as natural ingredients and extracts, have been shown to support gastrointestinal function and positively modulate gut microbiota composition [346,347,348,349,350,351].
Healthy dogs supplemented with oat groats, beet pulps, and pea fibers, prebiotics (i.e., inulin), probiotics, and immune supporters (i.e., spray-dried animal plasma and yeast-derived fermentation products), improved gastrointestinal and immune health, stool quality, and fecal metabolites [346]. Notably, a reduction in butyrate, isobutyrate, isovalerate, total branched-chain fatty acids (BCFA), indole, and ammonia fecal concentrations, and an increase in fecal 7-methylindole and calprotectin were observed. Similarly, the supplementation of Saccharomyces cerevisiae fermentation products positively shifted the gut microbiota composition in adult dogs, by increasing fecal Bifidobacterium and decreasing Fusobacterium, phenol, and indole concentrations [347]. Moreover, this prebiotic also enhanced the T helper 1 lymphocyte (Th1) response and decreased toll-like receptor (TLR) responsiveness, as well as the secretion of TNF-α and pro-inflammatory cytokines, demonstrating its potential anti-inflammatory and immune-modulating activity. Positive effects on gastrointestinal function and gut microbiota were also observed from the supplementation of other prebiotics, including miscanthus grass fiber [348], red ginseng dietary fiber [349], bioactive peptides [350], and Saccharomyces cerevisiae cell wall [351]. The latter, in particular, was responsible for a reduction in C. perfringens concentration in gut microbiota, thereby indirectly improving lactic acid concentration, which suggests an enhanced proliferation of beneficial lactic acid bacteria [351]. Prebiotics and probiotics also exert a beneficial effect on canine intestinal health by modulating the microflora through a direct competition for nutrients or the production of substances, such as bacteriocins, which inhibit the proliferation of other species, especially pathogens [352]. For example, the administration of Kefir, a fermented dairy product rich in lactic acid bacteria and beneficial yeasts, it has been shown to significantly reduce Clostridiaceae, Fusobacteriaceae, and Ruminococcaceae in healthy adult dogs while increasing Prevotellacea, Selenomonadaceae, Sutterellaceae, and lactic acid bacteria, thereby emerging as a novel functional pet food [353]. Finally, a favorable modulation of intestinal microbiota was attained in healthy dogs that were administered a snack containing 0.5% krill oil (KO). KO facilitated the proliferation of beneficial bacteria, including Bifidobacterium, Muribaculaceae, Ruminococcaceae, Faecalibacterium, and Prevotellaceae, thereby exerting a positive influence on the overall health of the canines [354].
Besides influencing gut microbiota composition, nutraceuticals can promote health and longevity by playing a role in the management of gastrointestinal diseases, accelerating the healing process, and the animals’ recovery. For example, a probiotic supplement containing four different Lactobacillus strains enhanced the multiplication of beneficial gut microbes to the detriment of potentially pathogenic ones, thereby promoting recovery in puppies with gastroenteritis [355]. Likewise, probiotic supplementation restored normobiosis in adult dogs suffering from diarrhea, thereby accelerating their recovery [356,357].
Dogs supplemented with a probiotic containing seven bacterial strains belonging to the genera Lactobacillus, Bifidobacterium, and Streptococcus showed faster recovery and accelerated normalization of the gut microflora compared to placebo control [192]. Similarly, supplementing dogs suffering from acute or intermittent diarrhea with a specific probiotic combination of Lactobacilli resulted in decreased concentrations of Clostridium perfringens and Enterococcus faecium along with normalization of stool consistency and improved well-being [358].
Currently, research is focusing on molecules derived from plants, such as turmeric, Boswellia serrata, or Aloe vera, whose bioactive substances, including curcumin, boswellic acid, vitamins, terpenoids, and flavonoids, have been proposed in the management of gastrointestinal disorders, such as colitis and IBD, through their anti-inflammatory and antioxidant properties. However, further research is necessary, particularly in the context of cats, to fully understand the potential of these substances [359].

3.6. Behavioral Disturbances and Cognitive Impairment Management by Nutraceuticals

Companion animals’ behavioral issues are a significant concern in the relationship between owners and pets [360]. Sharing everyday life with a pet that exhibits behavioral problems, such as aggression, destructive behaviors, vocalizations, or elimination issues, can cause stress in owners, undermine their overall well-being and social interactions with neighbors, as well as lead to serious consequences for the pet, including abandonment [361,362]. The onset of stress and behavioral disorders can be attributed to a variety of factors, including an imbalance of neurotransmitters like GABA, serotonin, dopamine, melatonin, histamine, acetylcholine, and norepinephrine [328,363]. These molecules have been linked to a range of neuropsychological processes, including anger, aggression, anxiety, motivation, reward, and emotional behaviors [328,364,365]. Consequently, an imbalance in their concentration has the potential to induce behavioral issues, as observed in dogs [366,367,368,369].
Growing evidence is supporting the relationship between gut microbiota and the behavior of companion animals through the gut–brain axis [196,360,370], possibly due to the activity of Lactobacilli, Bifidobacteria, and Escherichia coli, able to produce specific molecules, including neurotransmitters, which impact behavior and neurodevelopment [257,363].
In this sense, nutritional interventions have been proposed as a potential strategy to mitigate undesirable behaviors and enhance the quality of shared living [196].
Nutraceuticals, in particular probiotics [199,200], along with novel techniques, such as fecal microbiota transplantation (FMT), might represent a sustainable way to modulate gut microbiota and influence the GBA and, indirectly, pets’ behavior [371]. For example, Lactobacillus plantarum PS128 has been shown to improve general emotional stability in dogs while reducing the severity of behavioral problems, including aggression, separation anxiety, and compulsive disorders [199]. The dogs’ emotional and cognitive status, as well as behavioral issues and severity, were evaluated using the Evaluation of Dogs’ Emotional and Cognitive Disorders (EDED) scale and the Canine Behavioral Checklist (CBD) scale, respectively. Both scales’ values resulted decreased, thereby confirming the favorable impact of the probiotic on the animals’ behavior. In addition, a significant decrease in the serotonin turnover ratio (5-HIAA/5-HT) was observed in dogs suffering from separation anxiety, which was associated with a slowdown of serotonin metabolism after the PS128 supplementation.
Similarly, Lactobacillus plantarum LP185TM was observed to reduce aggression and anxiety, as indicated by the Canine Behavioral Assessment & Research Questionnaires (C-BARQ) [200]. The probiotic supplementation resulted in a decrease in the time required for the subjects to settle after the owner departure and in the dog daytime activity, and an improvement in their sleep patterns. These results suggest that supplementation with Lactobacillus plantarum LP185TM may be a promising strategy for treating behavioral issues.
Also, vegetal substances and extracts have been shown to modulate companion animals’ behavior. For instance, a 45-day administration of a nutraceutical diet containing Punica granatum, Valeriana officinalis, Rosmarinus officinalis, Tilia species, Crataegus oxyacantha, green tea extract, L-tryptophan, and an omega-3/6 in a ratio of 1:0.8 resulted effective in the management of behavioral issues (anxiety and chronic stress) in dogs [48]. Specifically, the animals receiving the nutraceutical diet showed increased plasma concentrations of serotonin, dopamine, and β-endorphins, together with decreased concentrations of noradrenaline and cortisol compared to the control group. Likewise, a similar nutraceutical supplementation to dogs with evident symptoms of behavioral disturbances was also linked to the improvement of conduct, such as anxiety, diffidence, irregular biorhythm, reactivity, irritability, and alertness, and their related clinical signs, including cutaneous and gastrointestinal signs [47]. The supplementation of a nutraceutical product containing anti-inflammatory compounds, pre- and probiotics, 5-hydroxytryptophan, and L-theanine alleviated stress and anxious behaviors in dogs, thus demonstrating the connection between nutraceuticals, microbiota, and behavior [372]. Finally, 20 healthy dogs receiving Melissa officinalis hydro-alcoholic extract showed a decrease in plasmatic 4-hydroxybutyric acid (GHB), one of the main metabolites synthesized from GABA [373]. The reduction was ascribed to the inhibitory activity of certain compounds present in the natural extract on GABA transaminase, which led to the accumulation of GABA in the brain and the manifestation of calming effect.
Behavioral disorders, such as disorientation, altered interactions with humans and other pets, and sleep–wake cycle disturbances, have also been observed in conjunction with cognitive impairment, in particular in elderly pets [374,375]. In this case, nutraceutical supplementation with Grifola frondosa, Curcuma longa, Carica papaya, Punica granatum, Aloe vera, Polygonum cuspidatum, Solanum lycopersicum, Vitis vinifera, Rosmarinus officinalis and an Omega 3/6 ratio of 1 : 0.8 showed to counteract the onset of cognitive impairment by increasing serum concentrations of brain-derived neurotrophic factor (BDNF), which is involved in neuroprotection [376].
Furthermore, the administration of Ginkgo biloba leaf extracts has been shown to ameliorate most of clinical indications associated with cognitive impairment, including disorientation, sleep disturbances, behavioral changes, and general physical condition in elderly dogs, suggesting its potential in enhancing their quality of life [377]. Also, nutraceutical supplements have been shown to exert beneficial effects on memory [378] and in the prevention of cognitive decline [379] in aged dogs. In cats, nutraceutical substances, such as fish oil, vitamins, antioxidants, minerals, botanicals, and amino acids, can help mitigate mild cognitive dysfunction syndrome (CDS) and enhance longevity [380].
It appears evident, then, that the evidence summarized here underscores the potential of nutraceuticals as a sustainable and effective strategy for the management of behavioral disorders and cognitive impairment in companion animals, ultimately supporting improvements in their welfare and longevity.

3.7. Joint Health Management by Nutraceuticals

Advancements in veterinary medicine and animal nutrition have contributed to an increase in the lifespan of companion animals [81]. Consequently, a greater number of animals are reaching advanced age and, consequently, developing age-related conditions, in particular OA [381], a widespread, degenerative, and inflammatory musculoskeletal disorder [382,383,384]. OA is characterized by a deterioration of the normal structure of the joint, which includes the degradation of articular cartilage, the remodeling of bone, the formation of osteophytes, subchondral bone sclerosis, chronic synovitis, and pain [214], which strongly reduces the quality of life of the affected animals [385].
In cats, OA may manifest with slight behavioral changes indicative of pain, rather than the conventional clinical signs observed in dogs (pain or tenderness, decreased range of motion, swelling, stiffness, muscle atrophy, crepitus, and effusion) [384]. A primary concern associated with OA is its incurability [214,383]. Consequently, treatment regimens should prioritize the mitigation of its progression, the reduction in pain, the improvement of motor function, and the enhancement of the quality of life for affected pets [214,383].
Although the presence of different approaches to managing this condition, including pharmacological [e.g., opioids or non-steroidal anti-inflammatory drugs (NSAIDs)] [386], dietary modifications and surgery [384,387], NSAIDs are the primary therapeutic choice.
However, the prolonged use of NSAIDs has been associated with adverse effects on various organs, including gastrointestinal erosion or ulceration, hepatic and renal damage, accelerated cartilage degeneration, and delayed bone healing [386,388]. For this reason, research has centered on identifying novel therapeutic options, such as nutraceuticals, which might constitute a more natural and safer alternative to NSAIDs [386]. Omega-3 fatty acids, glucosamine, chondroitin sulfate, collagen derivatives, green-lipped mussel, and various herbal medicines, including Boswellia serrata, were shown to enhance joint health and alleviate pain in dogs afflicted with OA [214,384,389,390]. For example, supplementing dogs with omega-3 fatty acids improved their joint conditions, reducing lameness and pain and improving their ability to rise from a resting position, walk, and play [141,391,392,393,394]. According to a study of Mehler et al. (2015), a daily administration of EPA and DHA in dogs suffering from OA significantly improved crepitus, pain, effusion, muscle atrophy and chronic nerve stimulation after 84 days [388]. Based on these observations, Corbee et al. (2013) found that 10 weeks of omega-3 fatty acids supplementation improved the behavior and locomotion of cats with OA [395]. Consequently, their activity level, movement, and interaction with their owners increased, and stiffness during gait decreased.
Furthermore, according to Fritsch et al. (2010), the supplementation of an omega-3-enriched diet in dogs suffering from OA and receiving the NSAID carprofen led to a reduction in its usage [140]. However, it should be noted that this study involved different veterinarians who adopted their own criteria to modulate the carprofen dosage. Therefore, this may have influenced the study’s findings. Other nutraceutical products proposed as potential treatments or adjuvants for OA include collagen and its derivatives, glucosamine, and plants or plant-derived supplements, such as Boswellia serrata, curcuminoids, and cannabidiol [214,396,397,398,399,400,401]. Recent findings have indicated that supplementing dogs with collagen hydrolysate or sulfated glucosamine, in comparison to a control diet, has led to a more significant reduction in lameness, OA-related symptoms, and pain.
Both of these nutraceuticals have been shown to enhance mobility, agility, and overall quality of life [396]. These outcomes were associated with the capacity of collagen hydrolysates to decrease the blood concentration of matrix metalloproteinase 3 (MMP-3), a proteolytic enzyme implicated in cartilage degradation [402], without affecting the levels of its inhibitor (Tissue Inhibitors of Metalloproteinases-1—TIMP-1). Moreover, the influence of both compounds on multiple biochemical processes has been postulated, with the potential to promote cartilage health [396]. This hypothesis suggests a positive impact on joint health, thereby substantiating their application in the management of OA.
In addition, beneficial effects on joint health have also been reported for collagen peptides [403,404]. In a study of dogs with OA, the administration of bioactive collagen peptides (BCP) via oral supplementation for 12 weeks yielded to superior outcomes in terms of alleviating the symptoms associated with OA when compared to other nutraceuticals (i.e., omega-3 fatty acids and Vitamin E) [398]. An improvement in clinical symptoms was also observed after oral supplementation with undenatured type II collagen (UC-II ®) in subjects suffering from OA [397]. In this study, metabolic alterations in the synovial fluid composition of joints in dogs suffering from OA were observed and compared to those in healthy controls. The alterations included increased levels of β-hydroxybutyrate, glutamine, trimethylamine-N-oxide (TMAO), creatine/creatinine, alanine, and histidine. The oral administration of UC-II resulted in a rebalancing of metabolism within the joint, as evidenced by the absence of β-hydroxybutyrate, a characteristic compound identified in the joints of dogs afflicted with OA.
As for plants and plant-derived substances, a substantial body of research has been conducted on their effects on OA, alone or in combination with other nutraceuticals [214,399,401,405,406]. Boswellia serrata is one of the most common nutraceuticals among those who support joint health. Boswellia-based products have produced positive results in various formulations. For example, a Boswellia serrata resin extract was shown to reduce OA symptoms in dogs that were not treated with any other anti-inflammatory agent after six weeks from the first day of administration [214]. Furthermore, the combination of Boswellia serrata and UC-II® improved mobility and synovial metabolomic composition in sick dogs’ joints after four weeks of administration, like the previous study [405]. In addition, Boswellia serrata is often an ingredient in nutraceutical blends that also contain other natural substances, such as chlorophyll, green tea extract, and natural chondroprotectants [406]. These blends may also contain fatty acids, whole freeze-dried green-lipped mussel powder (Perna canaliculus), and devil’s claw (Harpagophytum procumbens) [399]. These blends have been seen to reduce pain and clinical signs of OA and slow down the progression of the disease.
Although literature on this topic is extensive and opinions are sometimes conflicting, the reported evidence suggests that integrating nutraceuticals into multimodal management strategies is a scientifically substantiated, physiologically targeted approach to supporting joint integrity, enhancing longevity, and improving quality of life in companion animals.

3.8. Skin and Coat Management by Nutraceuticals

Skin is the largest organ of the body in mammals and represents their first line of defense [407]. It is strictly connected with the coat, both contributing to many fundamental functions, such as physical protection, thermoregulation, production of substances, including Vitamin D, and sensory perception [65,408]. The coat can also be considered a mirror of the overall health status, quality of life, physical and nutritional conditions of cats and dogs [409]. Ensuring an optimal healthy status of skin and coat is thereby pivotal to keeping their functions. Indeed, hair loss exposes skin to environmental stressors and may lead to the onset of diseases, such as dermatosis, thereby weakening its protective function.
Nutritional deficiencies have been shown to result in skin disorders, including seborrhea, keratinization, erythema, poor hair growth or alopecia, and greasy skin, which can facilitate the onset of pruritus and bacterial infections [410], thus potentially affecting the integrity of skin.
On the other hand, multiple nutraceuticals, including fatty acids [411,412], botanicals [42], probiotics [413,414], and trace minerals [415] can positively impact the skin and coat in both physiological and pathological conditions [409]. An increase in the total PUFAs content of a diet, from 9 to 13%, was shown to significantly enhance skin and coat quality, especially glossiness and softness of healthy dogs after 12 weeks supplementation, if compared to a control diet richer in saturated fatty acids [416].
The health status of the skin and coat also benefited from the administration of capsules containing EPA, DHA, and vitamin E. The observed benefits reached their zenith two months after the beginning of the supplementation and persisted for up to one month following its withdrawal [411].
KO constitutes an additional natural source of EPA and DHA. In a recent study conducted by Wang et al. (2025), the administration of a dietary supplement containing KO for 8 weeks in healthy dogs resulted in augmentation of total amino acids, particularly methionine, in the air of the supplemented subjects [354]. Furthermore, a decrease in thickness accompanied by an increase in hair softness was observed, thereby substantiating the capacity of KO to enhance the overall quality of canine hair.
The presence of trace minerals is of critical importance to the well-being and aesthetic quality of animals. Indeed, in a recent study by Amundson et al. (2025) the supplementation with zinc (Zn), manganese (Mn), copper (Cu), and iron (Fe) improved the hair quality, growth, and shedding, with more evident results in the dogs receiving acid-complexed organic trace mineral sources than in those receiving the standard inorganic source [415].
Probiotics, as well, have been observed to contribute to improving hair conditions in 5 healthy cats that received a probiotic supplement consisting of Bifidobacterium lactis and Lactobacillus plantarum for 28 days and were weekly assessed for body weight, overall physical condition, hair, and fecal [416]. Regarding hair quality, a statistically significant improvement was noted after 28 days of probiotic supplementation.
Supplementing a diet consisting of a blend of botanicals, zinc, and an omega-3/6 ratio of 1:0.8 to a dog with hind paw melanoma has been shown to reduce the lesion dimension and enhance the hematochemical profile and overall quality of life, thereby extending its median survival time [34]. Similarly, nutraceuticals exerted a favorable impact on a canine patient suffering from granulomatous dermatitis and chronic bilateral otitis. The dog was administered two nutraceutical diets for one month each. The first formulation, composed of fish proteins and potato carbohydrates, Rosa canina, Salvia officinalis, and Vaccinium macrocarpon, was tailored for cutaneous manifestations and resulted in a substantial decrease in the lesions associated with granulomatous dermatitis (i.e., papules, nodules, and plaques). Similarly, the second nutraceutical diet, based on fish proteins, rice carbohydrates, Melaleuca alternifolia, Tilia platyphyllos scapoli, and Tilia cordata, Allium sativum, Rosa canina, and zinc, was specific for auricular diseases and resulted in reduced inflammation, edema, erythema, and occlusion of the ear canal [55]. Similar results also emerged in a study conducted by Di Cerbo et al. (2016), where combining topical pharmacological treatment with nutraceutical supplementation for 90 days resulted in relieving the symptoms of chronic otitis externa, thereby improving the quality of life of these pets [46].
Nutraceuticals supplementation can result useful also in the management of allergic conditions in both cats and dogs. Gut microbiota modulation through probiotic administration, such as Lactobacillus sakei Probio-65, Bifidobacterium bifidum, Lactobacillus acidophilus, and Enterococcus faecium, improved all the symptoms related to canine atopic dermatitis (CAD), demonstrating the existence of a gut-skin axis [413,414,417]. Likewise, cats suffering from non–flea hypersensitivity dermatitis (NFHD), who were treated with methylprednisolone, exhibited reduced pruritus and a considerably prolonged time-to-relapse when receiving a co- and post-administration with the naturally occurring fatty acid palmitoylethanolamide compared to placebo [412].
In addition, the efficacy of nutraceuticals in mitigating the clinical symptoms and the severity of skin lesions in cats with cutaneous adverse food reaction (CAFR) has been well-documented [42,418,419,420]. CAFR is a group of reactions that can result from both allergies and intolerances. The administration of a nutraceutical diet based on Aloe vera, Arctium lappa, Malva sylvestris, Ribes nigrum, Allium sativum and Omega3/6 fatty acids (1:3 ratio), vitamin A, vitamin E, choline chloride, zinc sulphate monohydrate, cupric chelate glycine hydrate, and DL-methionine led to the restoration of physiologic dermal homeostasis, a reduction in the severity of the skin lesions, and an improvement in the clinical symptoms of CAFR, including drooling, pruritus, neck eczema, chronic conjunctivitis, and stomatitis [42].
In conclusion, these studies suggest the possible role of nutraceuticals in preserving the health and integrity of the skin and coat, thereby enhancing the quality of life and longevity in companion animals.

3.9. Immune Modulation by Nutraceuticals

As early as 1976, Scrimshaw hypothesized a link between nutrition and immune status [421]. Subsequent research has validated this association, and a great number of studies has been continuously increasing [422]. An imbalance or inadequate nutrition has a detrimental effect on immune function, increasing susceptibility to infections and compromising the organism’s capacity to repair damaged tissues or impeding the proliferation of malignant neoplastic cells [423]. In contrast, a favorable nutritional status, characterized by adequate intake of nutraceutical, particularly vitamins, trace elements, and omega-3 fatty acids, can support immune system function and positively impact health and longevity [424,425].
Particularly, supplementing healthy dogs with Chenpi powder (CPP) enhanced antioxidant and immunological functions [426]. Chenpi, the dry peel of Citrus reticulata cv. Chachiensis, contains a variety of beneficial compounds, including flavonoids and essential oils, which possess antioxidant, anti-inflammatory, and immune-modulating properties. The administration of CPP led to an enhancement in the activity of antioxidant enzymes, notably SOD, glutathione peroxidase (GSH-Px), and catalase (CAT), accompanied by a decline in serum malondialdehyde (MDA) concentration. Furthermore, a decrease in pro-inflammatory cytokines, such as IL-8 and TNF-α, and an increase in fecal secretory immunoglobulin A (SIgA) were observed. Similarly, an increase in SOD, GSH-Px, CAT, and Immunoglobulin G (IgG) serum content, along with a decrease in TNF-α, IL-8, and IL-1β was obtained after supplementing healthy dogs with KO [354], while oral supplementation of beta-glucans was shown to modulate the humoral immune response in healthy dogs, before and after vaccination [427]. Moreover, despite the absence of a significant impact on the white blood cell (WBC) count, the administration of dietary oat beta-glucans has been hypothesized to be implicated in the reduction in IL-4 serum concentration. This finding suggests a plausible mechanism through which these beta-glucans may hinder a T helper 2 lymphocyte (Th2) response following vaccination with the evaluated vaccine [428].
Also prebiotics, including GOS alone or in combination with manannoligosaccharides (MOS), FOS, and beta-glucans, enhanced immune function by increasing the number of polymorphonuclear cells, the phagocytosis index, and the ROS production for both Gram-positive and Gram-negative bacteria stimuli [429]. Similar results were also observed in beagle dogs after castration following the oral administration of a polysaccharide (from Astragalus membranaceus (APS). Besides positively affecting wound healing, the administration of APS resulted in a reduction in pro-inflammatory cytokines IL-1β and TNF-α, as well as C-reactive protein, a marker of inflammation, in plasma [430]. Concurrently, APS increased the serum content of IL-10.
Immunomodulatory properties were also associated with the supplementation of Ganoderma lucidum (15 mg/kg bw), which increased vaccine-specific IgG against rabies in serum, enhance the phagocytic activity of macrophages, and the percentage of major histocompatibility II (MHC-II) from B cells [431].
Another study reported that supplementation with Saccharomyces cerevisiae fermentation products favorably influenced Th1 response and diminished TLR responses, consequently leading to a reduction in inflammation in adult dogs [347]. Conversely, the administration of Saccharomyces cerevisiae cell wall to cats did not exert any influence on the immune system [351]. On the other hand, cats supplemented with diets containing yeast-derived nucleotides, salmon oil, or L-arginine improved immune system functionality. This was evidenced by enhanced lymphocyte proliferative responses to the T-cell mitogen phytohaemagglutinin and increased blood leukocyte phagocytic activity [432]. Finally, in cats, the simultaneous supplementation of Bifidobacterium lactis and Lactobacillus plantarum enhanced mucosal immunity by increasing IgA serum concentrations and improving antioxidant defense, specifically CAT, SOD, and GSH-PX. Moreover, these probiotics also showed to provide anti-inflammatory benefits by modulating cytokines involved in inflammation, such as TNF-α, IFN-γ, IL-4, and IL-2 [417]. Similarly, supplementing healthy dogs with a multi-strain probiotic resulted in beneficial modulation of fecal microbiota by reducing harmful bacteria, such as Clostridium perfringens, while increasing beneficial Bifidobacteria and Lactobacilli. An increase in fecal IgA and plasma IgG was also registered in the treated group [433].
The immune-modulating properties of nutraceuticals have also been assessed in the management of immune-mediated pathologies. An in vitro study conducted in 2016 showed the ability of several botanicals (Ascophyllum nodosum, Cucumis melo, Carica papaya, Aloe vera, Haematococcus pluvialis, Curcuma longa, Camellia sinensis, Punica granatum, Piper nigrum, Polygonum cuspidatum, Echinacea purpurea, Grifola frondosa, and Glycine max to modulate the production of proinflammatory cytokines, such as IFN-γ, in both human and canine peripheral blood mononucleated cells (PBMCs) [434]. Moreover, Haematococcus pluvialis, Glicine max, and the mixture of all the 13 botanicals hindered oxytetracycline (OTC)-induced toxicity. This is of relevance since OTC toxicity has been linked to detrimental effects on the immune system and the onset of immune-mediated diseases [435,436,437]. The same nutraceuticals, combined with conventional pharmacological treatment, resulted beneficial in the management of keratoconjunctivitis sicca (KCS), a multifactorial ocular pathology resulting from an immune dysregulation that leads to inflammatory alterations of the lacrimal gland and the subsequent deficiency in the tear aqueous fraction [44]. Besides ameliorating the clinical symptoms, such as blepharospasm, ocular hyperemia, periocular swelling, and ocular discharge, the combination reduced also conjunctival inflammation, corneal keratinization, corneal pigment density, mucus discharge, and tear production. Likewise, the same combination of nutraceuticals and conventional pharmacological treatment significantly improved the clinical conditions of dogs suffering from epiphora by modulating tear overflow, conjunctival inflammation, corneal keratinization, and blepharitis [45].
The immune-modulating activity of the aforementioned nutraceutical resulted also effective in the management of naturally infected dogs with Leishmania infantum [52]. The administration of nutraceutical supplements led to a restoration of platelet number and CD3+ CD4+ Foxp3+ Regulatory T (Treg) cells population, as well as an improvement in the CD4/CD8 ratio. Furthermore, the dogs receiving the nutraceutical diet exhibited a progressive decline in CD3+ CD4+ IFN-γ + Th1 cells, reaching levels comparable to those observed in the healthy control group by the end of the trial. Positive effects in the management of leishmaniosis were also associated with the oral administration of a supplement containing nucleotides and active hexose correlated compound (AHCC), which has been shown to possess immune-modulating properties [438]. Clinically healthy dogs infected with Leishmania infantum were treated with this nutraceutical supplement for 1 year. The treatment did not exert significant changes between the supplemented group and the placebo group in terms of CD4+ and CD8+ levels, CD4+/CD8+ ratio, cytokine levels, protein electrophoresis, complete blood count, biochemistry, parasite load, or chronic kidney disease staging. However, the ELISA test showed a significant reduction in serological titers of antibodies against Leishmania infantum, proving its ability to prevent disease progression, thereby protecting animals from becoming sick.
Given the sensitivity of immune cells to changes in the balance between oxidant and antioxidant species, another important factor to ensure optimal immune function is to maintain optimal antioxidant levels [439]. In this context, the administration of a natural antioxidant blend containing S-acetyl-glutathione (SAG), a precursor of glutathione originating from the fermentation of Saccharomyces cerevisiae, resulted in an increase in the glutathione peroxidase levels, a key enzyme against oxidative damage [440]. A summary of the action mechanism, the clinical outcome, the animal model and the contribution to longevity exerted by omega-3 fatty acids, prebiotics and probiotics, plant extracts and dietary supplements is provided in Table 1.

4. Conclusions

The shift in societal perception of pets as integral members of the family unit heightened the interest in their psychophysical health, well-being, and possibly longevity. Moreover, pet–owner interaction has been recognized as a mutually beneficial endeavor, where owner derives physical and psychological benefits and pet receives sustenance and protection. In addition, the love and caring provided both ways can contribute to the enhancement of their respective quality of life. Consequently, social interactions function as a preventative measure against the development of stress-related pathologies, which are frequently related to behavioral disturbs in both species.
The aforementioned shift is also concomitant with the greater sensitivity to the quality of food, nutritional requirements and habits that owners provide to their “new family members”. In this context, nutraceuticals can improve pet–owner relationships through a direct modulation of GBA, thus helpful in the prevention or management of behavioral disturbs.
Besides modulating the GBA, by regulating the production of neuromodulators, nutraceuticals can also positively impact the health of joints, skin, coat, and the immune system, thus representing a fundamental tool to ensure the well-being of pets, improve their quality of life, and possibly promote their longevity.

Author Contributions

Conceptualization, A.D.C.; methodology, A.D.C., M.N. and T.I.; software, A.D.C.; validation, A.D.C., M.N. and T.I.; formal analysis, A.D.C., M.N. and T.I.; investigation, A.D.C., M.N. and T.I.; resources, A.D.C.; data curation, M.N.; writing—original draft preparation, A.D.C., M.N. and T.I.; writing—review and editing, A.D.C., M.N. and T.I.; visualization, A.D.C., M.N. and T.I.; supervision, A.D.C. and T.I.; project administration, A.D.C.; funding acquisition, A.D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Kormotech LLC., Lviv, Lviv region, Ukraine (1411/2025).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
CRcaloric restriction
PUFAsPolyunsaturated fatty acids
SCshort-chain
ALAalpha-linolenic acid
LClong-chain
EPAEicosapentaenoic acid
DHAdocosahexaenoic acid
NSAIDnon-steroidal anti-inflammatory drug
AAarachidonic acid
GFRglomerular filtration rate
FOSfructooligosaccharides
GOSgalactooligosaccharides
XOSXylooligosaccharides
SCFAshort-chain fatty acids
GLP-1glucagon-like peptide-1
CKDchronic kidney disease
BUNblood urea nitrogen
GBAgut–brain axis
NOnitric oxide
ILinterleukin
COXcyclooxygenase
TNFtumor necrosis factor
SODsuperoxide dismutase
MBDmineral and bone disease
GSHreduced Glutathione
ROSreactive oxygen species
GSSGoxidized Glutathione
OSoxidative stress
ALTalanine transaminase
ASTalanine aminotransferase
ALPalkaline phosphatase
GGTgamma-glutamyl transferase
BILbilirubin
OAosteoarthritis
DAdopamine
5-HTserotonin
GABAgamma-aminobutyric acid
ENSenteric nervous system
CNScentral nervous system
ASDautism spectrum disorders
PDParkinson’s disease
ADAlzheimer’s disease
BCFAbranched-chain fatty acids
TLRtoll-like receptor
IBDinflammatory bowel disease
FMTfecal microbiota transplantation
CBDCanine Behavioral Checklist
C-BARQCanine Behavioral Assessment & Research Questionnaires
GHB4-hydroxybutyric acid
BDNFbrain-derived neurotrophic factor
CDScognitive dysfunction syndrome
MMPmetalloproteinase
TMAOtrimethylamine-N-oxide
KOKrill oil
NFHDnonflea hypersensitivity dermatitis
CADcanine atopic dermatitis
CAFRcutaneous adverse food reaction
CPPChenpi powder
SIgAsecretory immunoglobulin A
WBCwhite blood cell
MHC-IImajor histocompatibility II complex
OTCoxytetracycline
HCThematocrit
IMHAimmune-mediated anemia
KCSkeratoconjunctivitis sicca
SAGS-acetyl-glutathione
IgGImmunoglobulin G

References

  1. Calancea, B.-A.; Daina, S.; Macri, A. The science of snacks: A review of dog treats. Front. Anim. Sci. 2024, 5, 1440644. [Google Scholar] [CrossRef]
  2. Kępińska-Pacelik, J.; Biel, W.; Mizielińska, M.; Iwański, R. Chemical Composition and Palatability of Nutraceutical Dog Snacks. Appl. Sci. 2023, 13, 2806. [Google Scholar] [CrossRef]
  3. Ho, J.; Hussain, S.; Sparagano, O. Did the COVID-19 Pandemic Spark a Public Interest in Pet Adoption? Front. Vet. Sci. 2021, 8, 647308. [Google Scholar] [CrossRef]
  4. Barroso, C.; Fonseca, A.J.M.; Cabrita, A.R.J. Vitamins, Minerals and Phytonutrients as Modulators of Canine Immune Function: A Literature Review. Vet. Sci. 2024, 11, 655. [Google Scholar] [CrossRef]
  5. Overgaauw, P.A.M.; Vinke, C.M.; Hagen, M.; Lipman, L.J.A. A One Health Perspective on the Human-Companion Animal Relationship with Emphasis on Zoonotic Aspects. Int. J. Environ. Res. Public Health 2020, 17, 3789. [Google Scholar] [CrossRef] [PubMed]
  6. Mosteller, J. Animal-companion extremes and underlying consumer themes. J. Bus. Res. 2008, 61, 512–521. [Google Scholar] [CrossRef]
  7. Brittany, L.W. Insights-Driven Development of Humanized Foods for Pets. Meat Muscle Biol. 2022, 6, 1–12. [Google Scholar] [CrossRef]
  8. Schleicher, M.; Cash, S.B.; Freeman, L.M. Determinants of pet food purchasing decisions. Can. Vet. J. 2019, 60, 644–650. [Google Scholar]
  9. Boya, U.O.; Dotson, M.J.; Hyatt, E.M. A comparison of dog food choice criteria across dog owner segments: An exploratory study. Int. J. Consum. Stud. 2015, 39, 74–82. [Google Scholar] [CrossRef]
  10. Buchanan, R.L.; Baker, R.C.; Charlton, A.J.; Riviere, J.E.; Standaert, R. Pet food safety: A shared concern. Br. J. Nutr. 2011, 106 (Suppl. S1), S78–S84. [Google Scholar] [CrossRef]
  11. Bouma, E.M.C.; Dijkstra, A.; Arnt Rosa, S. Owner’s Anthropomorphic Perceptions of Cats’ and Dogs’ Abilities Are Related to the Social Role of Pets, Owners’ Relationship Behaviors, and Social Support. Animals 2023, 13, 3644. [Google Scholar] [CrossRef]
  12. Prata, J.C. Survey of Pet Owner Attitudes on Diet Choices and Feeding Practices for Their Pets in Portugal. Animals 2022, 12, 2775. [Google Scholar] [CrossRef]
  13. Michel, K.E.; Willoughby, K.N.; Abood, S.K.; Fascetti, A.J.; Fleeman, L.M.; Freeman, L.M.; Laflamme, D.P.; Bauer, C.; Kemp, B.L.E.; Doren, J.R.V. Attitudes of pet owners toward pet foods and feeding management of cats and dogs. J. Am. Vet. Med. Assoc. 2008, 233, 1699–1703. [Google Scholar] [CrossRef]
  14. Vinassa, M.; Vergnano, D.; Valle, E.; Giribaldi, M.; Nery, J.; Prola, L.; Bergero, D.; Schiavone, A. Profiling Italian cat and dog owners’ perceptions of pet food quality traits. BMC Vet. Res. 2020, 16, 131. [Google Scholar] [CrossRef] [PubMed]
  15. Nasri, H.; Baradaran, A.; Shirzad, H.; Rafieian-Kopaei, M. New concepts in nutraceuticals as alternative for pharmaceuticals. Int. J. Prev. Med. 2014, 5, 1487–1499. [Google Scholar] [PubMed]
  16. Ruiz-Cano, D.; Arnao, M.B. Beneficial Effects of Nutraceuticals, Especially Polyphenols on Canine Health. Pets 2024, 1, 228–254. [Google Scholar] [CrossRef]
  17. Ahmed, L.; Zagidullin, N. Editorial: Nutraceuticals in cardiovascular diseases and their associated risk conditions. Front. Cardiovasc. Med. 2024, 11, 1468355. [Google Scholar] [CrossRef]
  18. Makkar, R.; Behl, T.; Bungau, S.; Zengin, G.; Mehta, V.; Kumar, A.; Uddin, M.S.; Ashraf, G.M.; Abdel-Daim, M.M.; Arora, S.; et al. Nutraceuticals in Neurological Disorders. Int. J. Mol. Sci. 2020, 21, 4424. [Google Scholar] [CrossRef]
  19. Gonzalez-Sarrias, A.; Larrosa, M.; Garcia-Conesa, M.T.; Tomas-Barberan, F.A.; Espin, J.C. Nutraceuticals for older people: Facts, fictions and gaps in knowledge. Maturitas 2013, 75, 313–334. [Google Scholar] [CrossRef]
  20. Rai, R.H.; Goyal, R.K.; Singh, R.B.; Handjiev, S.; Singh, J.; Darlenska, T.H.; Smail, M.M.A. Chapter 43—Vitamins and minerals as nutraceuticals in cardiovascular diseases and other chronic diseases. In Functional Foods and Nutraceuticals in Metabolic and Non-Communicable Diseases; Singh, R.B., Watanabe, S., Isaza, A.A., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 651–670. [Google Scholar] [CrossRef]
  21. Ahmad, M.F.; Ahmad, F.A.; Ashraf, S.A.; Alsayegh, A.A.; Tabassum, F.; Bantun, F.; Elbandy, M.; Shama, E.; Uddin, S.; Khanam, A. Chapter ten—Microbial vitamins as nutraceuticals and their role as health-promoting agents. In Microbial Vitamins and Carotenoids in Food Biotechnology; Ashraf, S.A., Kuddus, M., Eds.; Academic Press: Cambridge, MA, USA, 2024; pp. 261–298. [Google Scholar] [CrossRef]
  22. Surana, K.R.; Ahire, E.D.; Patil, S.J.; Mahajan, S.K.; Patil, D.M.; Sonawane, D.D. Introduction to Nutraceutical Vitamins. In Vitamins as Nutraceuticals: Recent Advances and Applications; Ahire, E.D., Keservani, R.K., Surana, K.R., Singh, S., Kesharwani, R.K., Eds.; Scrivener Publishing LLC: Beverly, MA, USA, 2023; pp. 1–34. [Google Scholar] [CrossRef]
  23. Mišurcová, L.; Machů, L.; Orsavová, J. Chapter 29—Seaweed Minerals as Nutraceuticals. In Advances in Food and Nutrition Research; Kim, S.-K., Ed.; Academic Press: Cambridge, MA, USA, 2011; Volume 64, pp. 371–390. [Google Scholar]
  24. Desai, S.S.; Mane, V.K. Health Perspective of Nutraceutical Fatty Acids; (Omega-3 and Omega-6 Fatty Acids). In Nutraceutical Fatty Acids from Oleaginous Microalgae; Patel, A.K., Matsakas, L., Eds.; Scrivener Publishing LLC: Beverly, MA, USA, 2020; pp. 227–248. [Google Scholar] [CrossRef]
  25. Zhang, Z.; Li, X.; Sang, S.; McClements, D.J.; Chen, L.; Long, J.; Jiao, A.; Jin, Z.; Qiu, C. Polyphenols as Plant-Based Nutraceuticals: Health Effects, Encapsulation, Nano-Delivery, and Application. Foods 2022, 11, 2189. [Google Scholar] [CrossRef]
  26. Sahiner, M.; Yilmaz, A.S.; Gungor, B.; Ayoubi, Y.; Sahiner, N. Therapeutic and Nutraceutical Effects of Polyphenolics from Natural Sources. Molecules 2022, 27, 6225. [Google Scholar] [CrossRef]
  27. Vamanu, E. Polyphenolic Nutraceuticals to Combat Oxidative Stress Through Microbiota Modulation. Front. Pharmacol. 2019, 10, 492. [Google Scholar] [CrossRef] [PubMed]
  28. Prakash, D.; Gupta, C. Glucosinolates: The Phytochemicals of Nutraceutical Importance. J. Complement. Integr. Med. 2012, 9, 13. [Google Scholar] [CrossRef] [PubMed]
  29. Jahangeer, M.; Fatima, R.; Ashiq, M.; Basharat, A.; Qamar, S.A.; Bilal, M.; Iqbal, H. Therapeutic and Biomedical Potentialities of Terpenoids—A Review. J. Pure Appl. Microbiol. 2021, 15, 471–483. [Google Scholar] [CrossRef]
  30. Archana, O.; Nagadesi, P.K. Mushroom alkaloids as nutraceuticals, bioactive and medicinal properties: A preliminary review. Plant Sci. Today 2024, 11, 651–661. [Google Scholar] [CrossRef]
  31. Naveen, J.; Baskaran, V. Antidiabetic plant-derived nutraceuticals: A critical review. Eur. J. Nutr. 2018, 57, 1275–1299. [Google Scholar] [CrossRef]
  32. Calvani, M.; Pasha, A.; Favre, C. Nutraceutical Boom in Cancer: Inside the Labyrinth of Reactive Oxygen Species. Int. J. Mol. Sci. 2020, 21, 1936. [Google Scholar] [CrossRef]
  33. Maiuolo, J.; Gliozzi, M.; Carresi, C.; Musolino, V.; Oppedisano, F.; Scarano, F.; Nucera, S.; Scicchitano, M.; Bosco, F.; Macri, R.; et al. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021, 13, 3834. [Google Scholar] [CrossRef]
  34. Canello, S.; Guidetti, G.; Di Cerbo, A.; Cocco, R. A case of canine dermal melanoma: A nutraceutical approach. Int. J. Appl. Res. Vet. Med. 2018, 16, 117–121. [Google Scholar]
  35. Moss, J.W.; Ramji, D.P. Nutraceutical therapies for atherosclerosis. Nat. Rev. Cardiol. 2016, 13, 513–532. [Google Scholar] [CrossRef]
  36. Moss, J.W.E.; Williams, J.O.; Ramji, D.P. Nutraceuticals as therapeutic agents for atherosclerosis. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2018, 1864, 1562–1572. [Google Scholar] [CrossRef]
  37. Sosnowska, B.; Penson, P.; Banach, M. The role of nutraceuticals in the prevention of cardiovascular disease. Cardiovasc. Diagn. Ther. 2017, 7, S21–S31. [Google Scholar] [CrossRef] [PubMed]
  38. Garg, A.; Garg, S.; Singh, V.; Rai, G.; Prakash, P.; Mishra, N. Chapter 18—Role of nutraceuticals in neurological disease. In Microbiota-Gut-Brain Axis and CNS Disorders; Mishra, N., Kumar, A., Eds.; Academic Press: Cambridge, MA, USA, 2025; pp. 409–439. [Google Scholar] [CrossRef]
  39. DuBourdieu, D. Veterinary Nutraceuticals Stability Testing. In Nutraceuticals in Veterinary Medicine; Gupta, R.C., Srivastava, A., Lall, R., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 765–774. [Google Scholar] [CrossRef]
  40. Finno, C.J. Veterinary Pet Supplements and Nutraceuticals. Nutr. Today 2020, 55, 97–101. [Google Scholar] [CrossRef] [PubMed]
  41. Sankaranarayanan, A. Nutraceuticals in Equine Medicine. In Nutraceuticals in Veterinary Medicine; Springer: Berlin/Heidelberg, Germany, 2019; pp. 649–655. [Google Scholar] [CrossRef]
  42. Mazzeranghi, F.; Zanotti, C.; Di Cerbo, A.; Verstegen, J.P.; Cocco, R.; Guidetti, G.; Canello, S. Clinical efficacy of nutraceutical diet for cats with clinical signs of cutaneus adverse food reaction (CAFR). Pol. J. Vet. Sci. 2017, 20, 269–276. [Google Scholar] [CrossRef] [PubMed]
  43. Canello, S.; Guidetti, G.; Di Cerbo, A.; Cocco, R. A Successful Nutraceutical Approach to Manage an Elderly Dog Presenting a Focal Granulomatous Dermatitis with a Concomitant Chronic Otitis. Int. J. Appl. Res. Vet. Med. 2019, 17, 53–56. [Google Scholar]
  44. Destefanis, S.; Giretto, D.; Muscolo, M.C.; Di Cerbo, A.; Guidetti, G.; Canello, S.; Giovazzino, A.; Centenaro, S.; Terrazzano, G. Clinical evaluation of a nutraceutical diet as an adjuvant to pharmacological treatment in dogs affected by Keratoconjunctivitis sicca. BMC Vet. Res. 2016, 12, 214. [Google Scholar] [CrossRef]
  45. Destefanis, S.; Giretto, D.; Muscolo, M.C.; Centenaro, S.; Guidetti, G.; Canello, S. Clinical Evaluation of a Nutraceutical Diet as an Adjuvant to Pharmacological Treatment in Dogs Affected by Epiphora. Int. J. Appl. Res. Vet. Med. 2017, 15, 61–66. [Google Scholar]
  46. Di Cerbo, A.; Centenaro, S.; Beribe, F.; Laus, F.; Cerquetella, M.; Spaterna, A.; Guidetti, G.; Canello, S.; Terrazzano, G. Clinical evaluation of an antiinflammatory and antioxidant diet effect in 30 dogs affected by chronic otitis externa: Preliminary results. Vet. Res. Commun. 2016, 40, 29–38. [Google Scholar] [CrossRef]
  47. Di Cerbo, A.; Sechi, S.; Canello, S.; Guidetti, G.; Fiore, F.; Cocco, R. Behavioral Disturbances: An Innovative Approach to Monitor the Modulatory Effects of a Nutraceutical Diet. J. Vis. Exp. 2017, 119, e54878. [Google Scholar] [CrossRef]
  48. Sechi, S.; Di Cerbo, A.; Canello, S.; Guidetti, G.; Chiavolelli, F.; Fiore, F.; Cocco, R. Effects in dogs with behavioural disorders of a commercial nutraceutical diet on stress and neuroendocrine parameters. Vet. Rec. 2017, 180, 18. [Google Scholar] [CrossRef]
  49. Ciribe, F.; Panzarella, R.; Pisu, M.C.; Di Cerbo, A.; Guidetti, G.; Canello, S. Hypospermia Improvement in Dogs Fed on a Nutraceutical Diet. Sci. World J. 2018, 2018, 9520204. [Google Scholar] [CrossRef]
  50. Di Cerbo, A.; Guidetti, G.; Canello, S.; Cocco, R. A possible correlation between diet, serum oxytetracycline concentration, and onset of reproductive disturbances in bitches: Clinical observations and preliminary results. Turk. J. Vet. Anim. Sci. 2019, 43, 523–531. [Google Scholar] [CrossRef]
  51. Di Cerbo, A.; Iannitti, T.; Guidetti, G.; Centenaro, S.; Canello, S.; Cocco, R. A nutraceutical diet based on Lespedeza spp., Vaccinium macrocarpon and Taraxacum officinale improves spontaneous feline chronic kidney disease. Physiol. Rep. 2018, 6, e13737. [Google Scholar] [CrossRef] [PubMed]
  52. Cortese, L.; Annunziatella, M.; Palatucci, A.T.; Lanzilli, S.; Rubino, V.; Di Cerbo, A.; Centenaro, S.; Guidetti, G.; Canello, S.; Terrazzano, G. An immune-modulating diet increases the regulatory T cells and reduces T helper 1 inflammatory response in Leishmaniosis affected dogs treated with standard therapy. BMC Vet. Res. 2015, 11, 295. [Google Scholar] [CrossRef] [PubMed]
  53. Di Cerbo, A.; Pezzuto, F.; Canello, S.; Guidetti, G.; Palmieri, B. Therapeutic Effectiveness of a Dietary Supplement for Management of Halitosis in Dogs. J. Vis. Exp. 2015, 101, e52717. [Google Scholar] [CrossRef] [PubMed]
  54. Canello, S.; Guidetti, G.; Di Cerbo, A.; Scarano, A.; Cocco, R. Unraveling a commercial formula to relieve halitosis in dogs. Int. J. Appl. Res. Vet. Med. 2019, 17, 22–26. [Google Scholar]
  55. Canello, S.; Centenaro, S.; Guidetti, G. Nutraceutical approach for struvite uroliths management in cats. Int. J. Appl. Res. Vet. Med. 2017, 15, 19–25. [Google Scholar]
  56. Bauer, J.E. Evaluation of nutraceuticals, dietary supplements, and functional food ingredients for companion animals. J. Am. Vet. Med. Assoc. 2001, 218, 1755–1760. [Google Scholar] [CrossRef]
  57. Future, M.R. Pet Food Nutraceutical Market to Reach 10.5 USD Billion by 2035 with 4.88% CAGR by Increasing Awareness of Pet Health. 2025. Available online: https://www.einpresswire.com/article/808518673/pet-food-nutraceutical-market-to-reach-10-5-usd-billion-by-2035-with-4-88-cagr-by-increasing-awareness-of-pet-health#:~:text=Pet%20Food%20Nutraceutical%20Market%20was,period%20from%202025%20to%202035 (accessed on 28 May 2025).
  58. Elrod, S.M.; Hofmeister, E.H. Veterinarians’ attitudes towards use of nutraceuticals. Can. J. Vet. Res. 2019, 83, 291–297. [Google Scholar]
  59. McNicholas, J.; Gilbey, A.; Rennie, A.; Ahmedzai, S.; Dono, J.A.; Ormerod, E. Pet ownership and human health: A brief review of evidence and issues. BMJ 2005, 331, 1252–1254. [Google Scholar] [CrossRef]
  60. Monks, S.; Clark, A. The role of pets in the lives of people with dementia: A scoping review. Aging Ment. Health 2024, 28, 1419–1426. [Google Scholar] [CrossRef]
  61. Health, N.i. Health Benefits of Human-Animal Interactions. 2018. Available online: https://newsinhealth.nih.gov/2018/02/power-pets (accessed on 6 June 2025).
  62. Hodgson, K.; Darling, M. Zooeyia: An essential component of “One Health”. Can. Vet. J. 2011, 52, 189–191. [Google Scholar]
  63. Wensley, S.P. Animal welfare and the human-animal bond: Considerations for veterinary faculty, students, and practitioners. J. Vet. Med. Educ. 2008, 35, 532–539. [Google Scholar] [CrossRef] [PubMed]
  64. Kienzle, E.; Bergler, R.; Mandernach, A. A comparison of the feeding behavior and the human-animal relationship in owners of normal and obese dogs. J. Nutr. 1998, 128, 2779S–2782S. [Google Scholar] [CrossRef] [PubMed]
  65. Downes, M.J.; Devitt, C.; Downes, M.T.; More, S.J. Understanding the context for pet cat and dog feeding and exercising behaviour among pet owners in Ireland: A qualitative study. Ir. Vet. J. 2017, 70, 29. [Google Scholar] [CrossRef] [PubMed]
  66. Hennessy, M.B.; Voith, V.L.; Young, T.L.; Hawke, J.L.; Centrone, J.; McDowell, A.L.; Linden, F.; Davenport, G.M. Exploring Human Interaction and Diet Effects on the Behavior of Dogs in a Public Animal Shelter. J. Appl. Anim. Welf. Sci. 2002, 5, 253–273. [Google Scholar] [CrossRef]
  67. Nielson, S.A.; Khosa, D.K.; Verbrugghe, A.; Clow, K.M. Talking treats: A qualitative study to understand the importance of treats in the pet-caregiver relationship. Prev. Vet. Med. 2024, 226, 106163. [Google Scholar] [CrossRef]
  68. Delgado, M.; Dantas, L.M.S. Feeding Cats for Optimal Mental and Behavioral Well-Being. Vet. Clin. N. Am. Small Anim. Pract. 2020, 50, 939–953. [Google Scholar] [CrossRef]
  69. Rawlings, J.M.; Culham, N. Halitosis in dogs and the effect of periodontal therapy. J. Nutr. 1998, 128, 2715S–2716S. [Google Scholar] [CrossRef]
  70. Eaton, R.; Emmas, S.-A.; Whelan, F.; Groom, A. A randomised, double-blind, placebo-controlled trial, assessing the effect of a nutraceutical tablet in the management of stress in pet dogs. Appl. Anim. Behav. Sci. 2021, 242, 105416. [Google Scholar] [CrossRef]
  71. Stott, G.H. What is Animal Stress and How is it Measured? J. Anim. Sci. 1981, 52, 150–153. [Google Scholar] [CrossRef]
  72. Fan, Z.; Bian, Z.; Huang, H.; Liu, T.; Ren, R.; Chen, X.; Zhang, X.; Wang, Y.; Deng, B.; Zhang, L. Dietary Strategies for Relieving Stress in Pet Dogs and Cats. Antioxidants 2023, 12, 545. [Google Scholar] [CrossRef]
  73. Beerda, B.; Schilder, M.B.H.; van Hooff, J.A.R.A.M.; de Vries, H.W.; Mol, J.A. Behavioural, saliva cortisol and heart rate responses to different types of stimuli in dogs. Appl. Anim. Behav. Sci. 1998, 58, 365–381. [Google Scholar] [CrossRef]
  74. Buller, K.; Ballantyne, K.C. Living with and loving a pet with behavioral problems: Pet owners’ experiences. J. Vet. Behav. 2020, 37, 41–47. [Google Scholar] [CrossRef]
  75. Adams, V.J.; Watson, P.; Carmichael, S.; Gerry, S.; Penell, J.; Morgan, D.M. Exceptional longevity and potential determinants of successful ageing in a cohort of 39 Labrador retrievers: Results of a prospective longitudinal study. Acta Vet. Scand. 2016, 58, 29. [Google Scholar] [CrossRef] [PubMed]
  76. Guo, X.; Wang, Y.; Zhu, Z.; Li, L. The Role of Plant Extracts in Enhancing Nutrition and Health for Dogs and Cats: Safety, Benefits, and Applications. Vet. Sci. 2024, 11, 426. [Google Scholar] [CrossRef]
  77. Butterwick, R.F. Impact of nutrition on ageing the process. Bridging the gap: The animal perspective. Br. J. Nutr. 2015, 113, S23–S25. [Google Scholar] [CrossRef]
  78. Fadnes, L.T.; Celis-Morales, C.; Økland, J.-M.; Parra-Soto, S.; Livingstone, K.M.; Ho, F.K.; Pell, J.P.; Balakrishna, R.; Javadi Arjmand, E.; Johansson, K.A.; et al. Life expectancy can increase by up to 10 years following sustained shifts towards healthier diets in the United Kingdom. Nat. Food 2023, 4, 961–965. [Google Scholar] [CrossRef]
  79. Hu, F.B. Diet strategies for promoting healthy aging and longevity: An epidemiological perspective. J. Int. Med. 2024, 295, 508–531. [Google Scholar] [CrossRef]
  80. Lo, W.C.; Hu, T.H.; Shih, C.Y.; Lin, H.H.; Hwang, J.S. Impact of Healthy Lifestyle Factors on Life Expectancy and Lifetime Health Care Expenditure: Nationwide Cohort Study. JMIR Public Health Surveill. 2024, 10, e57045. [Google Scholar] [CrossRef]
  81. Taylor, E.J.; Adams, C.; Neville, R. Some nutritional aspects of ageing in dogs and cats. Proc. Nutr. Soc. 1995, 54, 645–656. [Google Scholar] [CrossRef] [PubMed]
  82. Blanchard, T.; Mugnier, A.; Boulet, F.; Meynadier, A.; Priymenko, N. Epidemiological and clinical profiles of young and senior dogs fed a standard diet. Prev. Vet. Med. 2025, 240, 106537. [Google Scholar] [CrossRef]
  83. Kraft, W. Geriatrics in canine and feline internal medicine. Eur. J. Med. Res. 1998, 3, 31–41. [Google Scholar] [PubMed]
  84. Bontempo, V. Nutrition and Health of Dogs and Cats: Evolution of Petfood. Vet. Res. Commun. 2005, 29, 45–50. [Google Scholar] [CrossRef] [PubMed]
  85. Sparkes, A.H. Feeding old cats--an update on new nutritional therapies. Top. Companion. Anim. Med. 2011, 26, 37–42. [Google Scholar] [CrossRef]
  86. Hall, J.A.; Yerramilli, M.; Obare, E.; Yerramilli, M.; Panickar, K.S.; Bobe, G.; Jewell, D.E. Nutritional interventions that slow the age-associated decline in renal function in a canine geriatric model for elderly humans. J. Nutr. Health Aging 2016, 20, 1010–1023. [Google Scholar] [CrossRef]
  87. Haake, J.; Meyerhoff, N.; Meller, S.; Twele, F.; Charalambous, M.; Wilke, V.; Volk, H. Investigating Owner Use of Dietary Supplements in Dogs with Canine Cognitive Dysfunction. Animals 2023, 13, 3056. [Google Scholar] [CrossRef]
  88. Kealy, R.D.; Lawler, D.F.; Ballam, J.M.; Mantz, S.L.; Biery, D.N.; Greeley, E.H.; Lust, G.; Segre, M.; Smith, G.K.; Stowe, H.D. Effects of diet restriction on life span and age-related changes in dogs. J. Am. Vet. Med. Assoc. 2002, 220, 1315–1320. [Google Scholar] [CrossRef]
  89. Trepanowski, J.F.; Canale, R.E.; Marshall, K.E.; Kabir, M.M.; Bloomer, R.J. Impact of caloric and dietary restriction regimens on markers of health and longevity in humans and animals: A summary of available findings. Nutr. J. 2011, 10, 107. [Google Scholar] [CrossRef]
  90. Lawler, D.F.; Larson, B.T.; Ballam, J.M.; Smith, G.K.; Biery, D.N.; Evans, R.H.; Greeley, E.H.; Segre, M.; Stowe, H.D.; Kealy, R.D. Diet restriction and ageing in the dog: Major observations over two decades. Br. J. Nutr. 2008, 99, 793–805. [Google Scholar] [CrossRef]
  91. Vendelbo, M.H.; Nair, K.S. Mitochondrial longevity pathways. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2011, 1813, 634–644. [Google Scholar] [CrossRef] [PubMed]
  92. Carrillo, A.E.; Flouris, A.D. Caloric restriction and longevity: Effects of reduced body temperature. Ageing Res. Rev. 2011, 10, 153–162. [Google Scholar] [CrossRef] [PubMed]
  93. Watroba, M.; Szukiewicz, D. Sirtuins at the Service of Healthy Longevity. Front. Physiol. 2021, 12, 724506. [Google Scholar] [CrossRef] [PubMed]
  94. Pavlović, I.; Khatib, S.; Milisav, I.; Mahajna, J. Nutraceuticals for Promoting Longevity. Curr. Nutraceuticals 2020, 1, 18–32. [Google Scholar] [CrossRef]
  95. Rees, C.A.; Bauer, J.E.; Burkholder, W.J.; Kennis, R.A.; Dunbar, B.L.; Bigley, C.E. Effects of dietary flax seed and sunflower seed supplementation on normal canine serum polyunsaturated fatty acids and skin and hair coat condition scores. Vet. Dermatol. 2001, 12, 111–117. [Google Scholar] [CrossRef]
  96. Yoon, D.; Kim, Y.J.; Lee, W.K.; Choi, B.R.; Oh, S.M.; Lee, Y.S.; Kim, J.K.; Lee, D.Y. Metabolic Changes in Serum Metabolome of Beagle Dogs Fed Black Ginseng. Metabolites 2020, 10, 517. [Google Scholar] [CrossRef]
  97. Kim, Y.J.; Lee, D.Y.; Park, H.-E.; Yoon, D.; Lee, B.; Kim, J.G.; Im, K.-H.; Lee, Y.-S.; Lee, W.-K.; Kim, J.K. Serum Metabolic Profiling Reveals Potential Anti-Inflammatory Effects of the Intake of Black Ginseng Extracts in Beagle Dogs. Molecules 2020, 25, 3759. [Google Scholar] [CrossRef]
  98. Pinna, C.; Giuditta, V.C.; Vladimiro, C.; Teresa, R.-E.M.; Claudio, S.; Monica, G.; Paolo, G.P.; Biagi, G. An in vitro evaluation of the effects of a Yucca schidigera extract and chestnut tannins on composition and metabolic profiles of canine and feline faecal microbiota. Arch. Anim. Nutr. 2017, 71, 395–412. [Google Scholar] [CrossRef]
  99. Barry, K.A.; Hernot, D.C.; Middelbos, I.S.; Francis, C.; Dunsford, B.; Swanson, K.S.; Fahey, G.C., Jr. Low-level fructan supplementation of dogs enhances nutrient digestion and modifies stool metabolite concentrations, but does not alter fecal microbiota populations. J. Anim. Sci. 2009, 87, 3244–3252. [Google Scholar] [CrossRef]
  100. Soares, N.M.M.; Bastos, T.S.; Kaelle, G.C.B.; de Souza, R.B.M.d.S.; de Oliveira, S.G.; Félix, A.P. Digestibility and Palatability of the Diet and Intestinal Functionality of Dogs Fed a Blend of Yeast Cell Wall and Oregano Essential Oil. Animals 2023, 13, 2527. [Google Scholar] [CrossRef]
  101. Campigotto, G.; Alba, D.F.; Sulzbach, M.M.; Dos Santos, D.S.; Souza, C.F.; Baldissera, M.D.; Gundel, S.; Ourique, A.F.; Zimmer, F.; Petrolli, T.G.; et al. Dog food production using curcumin as antioxidant: Effects of intake on animal growth, health and feed conservation. Arch. Anim. Nutr. 2020, 74, 397–413. [Google Scholar] [CrossRef]
  102. Sgorlon, S.; Stefanon, B.; Sandri, M.; Colitti, M. Nutrigenomic activity of plant derived compounds in health and disease: Results of a dietary intervention study in dog. Res. Vet. Sci. 2016, 109, 142–148. [Google Scholar] [CrossRef]
  103. Baumgartner-Parzer, S.M.; Waldenberger, F.R.; Freudenthaler, A.; Ginouvès-Guerdoux, A.; McGahie, D.; Gatto, H. The Natural Antioxidants, Pomegranate Extract and Soy Isoflavones, Favourably Modulate Canine Endothelial Cell Function. Int. Sch. Res. Not. 2012, 2012, 590328. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, M.; Xue, J.; Li, Y.; Zhang, W.; Ma, D.; Liu, L.; Zhang, Z. Resveratrol protects against arsenic trioxide-induced nephrotoxicity by facilitating arsenic metabolism and decreasing oxidative stress. Arch. Toxicol. 2013, 87, 1025–1035. [Google Scholar] [CrossRef] [PubMed]
  105. Kobayashi, M.; Okada, Y.; Ueno, H.; Mizorogi, T.; Ohara, K.; Kawasumi, K.; Suruga, K.; Kadokura, K.; Ohnishi, Y.; Arai, T. Effects of Supplementation with Anti-Inflammatory Compound Extracted from Herbs in Healthy and Obese Cats. Vet. Med. 2020, 11, 39–44. [Google Scholar] [CrossRef]
  106. Rahman, S.U.; Huang, Y.; Zhu, L.; Chu, X.; Junejo, S.A.; Zhang, Y.; Khan, I.M.; Li, Y.; Feng, S.; Wu, J.; et al. Tea polyphenols attenuate liver inflammation by modulating obesity-related genes and down-regulating COX-2 and iNOS expression in high fat-fed dogs. BMC Vet. Res. 2020, 16, 234. [Google Scholar] [CrossRef] [PubMed]
  107. Fu, J.-h.; Zheng, Y.-q.; Li, P.; Li, X.-z.; Shang, X.-h.; Liu, J.-x. Hawthorn leaves flavonoids decreases inflammation related to acute myocardial ischemia/reperfusion in anesthetized dogs. Chin. J. Integr. Med. 2013, 19, 582–588. [Google Scholar] [CrossRef]
  108. Zhang, M.; Mo, R.; Wang, H.; Liu, T.; Zhang, G.; Wu, Y. Grape seed proanthocyanidin improves intestinal inflammation in canine through regulating gut microbiota and bile acid compositions. FASEB J. 2023, 37, e23285. [Google Scholar] [CrossRef]
  109. Gantt, W.H.; Newton, J.E.; Royer, F.L.; Stephens, J.H. Effect of person. 1966. Integr. Physiol. Behav. Sci. 1991, 26, 145–160. [Google Scholar] [CrossRef]
  110. Lynch, J.J.; McCarthy, J.F. Social responding in dogs: Heart rate changes to a person. Psychophysiology 1969, 5, 389–393. [Google Scholar] [CrossRef]
  111. Mârza, S.M.; Munteanu, C.; Papuc, I.; Radu, L.; Diana, P.; Purdoiu, R.C. Behavioral, Physiological, and Pathological Approaches of Cortisol in Dogs. Animals 2024, 14, 3536. [Google Scholar] [CrossRef] [PubMed]
  112. Csoltova, E.; Martineau, M.; Boissy, A.; Gilbert, C. Behavioral and physiological reactions in dogs to a veterinary examination: Owner-dog interactions improve canine well-being. Physiol. Behav. 2017, 177, 270–281. [Google Scholar] [CrossRef] [PubMed]
  113. Bergamasco, L.; Osella, M.C.; Savarino, P.; Larosa, G.; Ozella, L.; Manassero, M.; Badino, P.; Odore, R.; Barbero, R.; Re, G. Heart rate variability and saliva cortisol assessment in shelter dog: Human–animal interaction effects. Appl. Anim. Behav. Sci. 2010, 125, 56–68. [Google Scholar] [CrossRef]
  114. Shiverdecker, M.D.; Schiml, P.A.; Hennessy, M.B. Human interaction moderates plasma cortisol and behavioral responses of dogs to shelter housing. Physiol. Behav. 2013, 109, 75–79. [Google Scholar] [CrossRef]
  115. Thielke, L.E.; Udell, M.A. The role of oxytocin in relationships between dogs and humans and potential applications for the treatment of separation anxiety in dogs. Biol. Rev. Camb. Philos. Soc. 2017, 92, 378–388. [Google Scholar] [CrossRef]
  116. Odendaal, J.S.; Meintjes, R.A. Neurophysiological correlates of affiliative behaviour between humans and dogs. Vet. J. 2003, 165, 296–301. [Google Scholar] [CrossRef]
  117. Handlin, L.; Hydbring-Sandberg, E.; Nilsson, A.; Ejdebäck, M.; Jansson, A.; Uvnäs-Moberg, K. Short-Term Interaction between Dogs and Their Owners: Effects on Oxytocin, Cortisol, Insulin and Heart Rate—An Exploratory Study. Anthrozoös 2011, 24, 301–315. [Google Scholar] [CrossRef]
  118. Handlin, L.; Nilsson, A.; Ejdebäck, M.; Hydbring-Sandberg, E.; Uvnas-Moberg, K. Associations between the psychological characteristics of the human-dog relationship and oxytocin and cortisol levels. Anthrozoös 2012, 25, 215–228. [Google Scholar] [CrossRef]
  119. Rehnberg, L.K.; Robert, K.A.; Watson, S.J.; Peters, R.A. The effects of social interaction and environmental enrichment on the space use, behaviour and stress of owned housecats facing a novel environment. Appl. Anim. Behav. Sci. 2015, 169, 51–61. [Google Scholar] [CrossRef]
  120. Vitale, K.R.; Frank, D.H.; Conroy, J.; Udell, M.A.R. Cat Foster Program Outcomes: Behavior, Stress, and Cat–Human Interaction. Animals 2022, 12, 2166. [Google Scholar] [CrossRef]
  121. Gourkow, N.; Hamon, S.C.; Phillips, C.J.C. Effect of gentle stroking and vocalization on behaviour, mucosal immunity and upper respiratory disease in anxious shelter cats. Prev. Vet. Med. 2014, 117, 266–275. [Google Scholar] [CrossRef] [PubMed]
  122. National Institute of Health. Omega-3 Fatty Acids. 2024. Available online: https://ods.od.nih.gov/factsheets/Omega3FattyAcids-HealthProfessional/ (accessed on 15 June 2025).
  123. Abhari, K.; Mousavi Khaneghah, A. Alternative extraction techniques to obtain, isolate and purify proteins and bioactive from aquaculture and by-products. Adv. Food Nutr. Res. 2020, 92, 35–52. [Google Scholar] [CrossRef] [PubMed]
  124. Takic, M.; Pokimica, B.; Petrovic-Oggiano, G.; Popovic, T. Effects of Dietary α-Linolenic Acid Treatment and the Efficiency of Its Conversion to Eicosapentaenoic and Docosahexaenoic Acids in Obesity and Related Diseases. Molecules 2022, 27, 4471. [Google Scholar] [CrossRef] [PubMed]
  125. Rajaram, S. Health benefits of plant-derived α-linolenic acid123. Am. J. Clin. Nutr. 2014, 100, 443S–448S. [Google Scholar] [CrossRef]
  126. Kranz, S.; Huss, L.; Dobbs-Oates, J. Food Sources of EPA and DHA in the Diets of American Children, NHANES 2003–2010. BAOJ Nutr. 2015, 1, 5. [Google Scholar] [CrossRef]
  127. Brenna, J.T.; Salem, N., Jr.; Sinclair, A.J.; Cunnane, S.C. α-Linolenic acid supplementation and conversion to n-3 long-chain polyunsaturated fatty acids in humans. Prostaglandins Leukot. Essent. Fat. Acids 2009, 80, 85–91. [Google Scholar] [CrossRef]
  128. Stoeckel, K.; Nielsen, L.H.; Fuhrmann, H.; Bachmann, L. Fatty acid patterns of dog erythrocyte membranes after feeding of a fish-oil based DHA-rich supplement with a base diet low in n-3 fatty acids versus a diet containing added n-3 fatty acids. Acta Vet. Scand. 2011, 53, 57. [Google Scholar] [CrossRef]
  129. Lenox, C.E.; Bauer, J.E. Potential Adverse Effects of Omega-3 Fatty Acids in Dogs and Cats. J. Vet. Int. Med. 2013, 27, 217–226. [Google Scholar] [CrossRef]
  130. Bauer, J.E. Responses of dogs to dietary omega-3 fatty acids. J. Am. Vet. Med. Assoc. 2007, 231, 1657–1661. [Google Scholar] [CrossRef]
  131. Bauer, J.E. Metabolic basis for the essential nature of fatty acids and the unique dietary fatty acid requirements of cats. J. Am. Vet. Med. Assoc. 2006, 229, 1729–1732. [Google Scholar] [CrossRef]
  132. Masmeijer, C.; van Leenen, K.; De Cremer, L.; Deprez, P.; Cox, E.; Devriendt, B.; Pardon, B. Effects of omega-3 fatty acids on immune, health and growth variables in veal calves. Prev. Vet. Med. 2020, 179, 104979. [Google Scholar] [CrossRef] [PubMed]
  133. Yaqoob, P.; Calder, P. Effects of dietary lipid manipulation upon inflammatory mediator production by murine macrophages. Cell Immunol. 1995, 163, 120–128. [Google Scholar] [CrossRef] [PubMed]
  134. Baker, E.J.; Yusof, M.H.; Yaqoob, P.; Miles, E.A.; Calder, P.C. Omega-3 fatty acids and leukocyte-endothelium adhesion: Novel anti-atherosclerotic actions. Mol. Asp. Med. 2018, 64, 169–181. [Google Scholar] [CrossRef] [PubMed]
  135. De Caterina, R.; Cybulsky, M.A.; Clinton, S.K.; Gimbrone, M.A., Jr.; Libby, P. Omega-3 fatty acids and endothelial leukocyte adhesion molecules. Prostaglandins Leukot. Essent. Fat. Acids 1995, 52, 191–195. [Google Scholar] [CrossRef]
  136. Hughes, D.A.; Southon, S.; Pinder, A.C. (n-3) Polyunsaturated Fatty Acids Modulate the Expression of Functionally Associated Molecules on Human Monocytes in Vitro1. J. Nutr. 1996, 126, 603–610. [Google Scholar] [CrossRef]
  137. Miles, E.A.; Wallace, F.A.; Calder, P.C. Dietary fish oil reduces intercellular adhesion molecule 1 and scavenger receptor expression on murine macrophages. Atherosclerosis 2000, 152, 43–50. [Google Scholar] [CrossRef]
  138. Sanderson, P.; Calder, P.C. Dietary fish oil diminishes lymphocyte adhesion to macrophage and endothelial cell monolayers. Immunology 1998, 94, 79–87. [Google Scholar] [CrossRef]
  139. Giacobbe, J.; Benoiton, B.; Zunszain, P.; Pariante, C.M.; Borsini, A. The Anti-Inflammatory Role of Omega-3 Polyunsaturated Fatty Acids Metabolites in Pre-Clinical Models of Psychiatric, Neurodegenerative, and Neurological Disorders. Front. Psychiatry 2020, 11, 122. [Google Scholar] [CrossRef]
  140. Fritsch, D.A.; Allen, T.A.; Dodd, C.E.; Jewell, D.E.; Sixby, K.A.; Leventhal, P.S.; Brejda, J.; Hahn, K.A. A multicenter study of the effect of dietary supplementation with fish oil omega-3 fatty acids on carprofen dosage in dogs with osteoarthritis. J. Am. Vet. Med. Assoc. 2010, 236, 535–539. [Google Scholar] [CrossRef]
  141. Roush, J.K.; Dodd, C.E.; Fritsch, D.A.; Allen, T.A.; Jewell, D.E.; Schoenherr, W.D.; Richardson, D.C.; Leventhal, P.S.; Hahn, K.A. Multicenter veterinary practice assessment of the effects of omega-3 fatty acids on osteoarthritis in dogs. J. Am. Vet. Med. Assoc. 2010, 236, 59–66. [Google Scholar] [CrossRef]
  142. Richards, T.L.; Burron, S.; Ma, D.W.L.; Pearson, W.; Trevizan, L.; Minikhiem, D.; Grant, C.; Patterson, K.; Shoveller, A.K. Effects of dietary camelina, flaxseed, and canola oil supplementation on inflammatory and oxidative markers, transepidermal water loss, and coat quality in healthy adult dogs. Front. Vet. Sci. 2023, 10, 1085890. [Google Scholar] [CrossRef]
  143. Logas, D.; Kunkle, G.A. Double-blinded Crossover Study with Marine Oil Supplementation Containing High-dose icosapentaenoic Acid for the Treatment of Canine Pruritic Skin Disease. Vet. Dermatol. 1994, 5, 99–104. [Google Scholar] [CrossRef]
  144. Mueller, R.S.; Fieseler, K.V.; Fettman, M.J.; Zabel, S.; Rosychuk, R.A.W.; Ogilvie, G.K.; Greenwalt, T.L. Effect of omega-3 fatty acids on canine atopic dermatitis. J. Small Anim. Pract. 2004, 45, 293–297. [Google Scholar] [CrossRef]
  145. Zhang, Z.-X.; Lin, Y.-C.; Lian, M.; Li, Y.-F.; Chen, J.-F.; Ma, X.-L.; Guo, D.-S.; Yang, G.; Sun, X.-M. Evaluation of the efficacy and safety of omega-3 fatty acid nutritional supplements from Schizochytrium sp. in dog food. Algal. Res. 2025, 89, 104072. [Google Scholar] [CrossRef]
  146. Billman, G.E.; Kang, J.X.; Leaf, A. Prevention of ischemia-induced cardiac Sudden death by n−3 polyunsaturated fatty acids in dogs. Lipids 1997, 32, 1161–1168. [Google Scholar] [CrossRef] [PubMed]
  147. Leaf, A. The electrophysiologic basis for the antiarrhythmic and anticonvulsant effects of n−3 polyunsaturated fatty acids: Heart and brain. Lipids 2001, 36, S107–S110. [Google Scholar] [CrossRef] [PubMed]
  148. Hock, C.E.; Beck, L.D.; Bodine, R.C.; Reibel, D.K. Influence of dietary n-3 fatty acids on myocardial ischemia and reperfusion. Am. J. Physiol. Heart Circ. Physiol. 1990, 259, H1518–H1526. [Google Scholar] [CrossRef]
  149. Quattrone, A.; Belabbas, R.; Fehri, N.E.; Agradi, S.; Mazzola, S.M.; Barbato, O.; Dal Bosco, A.; Mattioli, S.; Failla, S.; Abdel-Kafy, E.-S.M.; et al. The Effect of Dietary Plant-Derived Omega 3 Fatty Acids on the Reproductive Performance and Gastrointestinal Health of Female Rabbits. Vet. Sci. 2024, 11, 457. [Google Scholar] [CrossRef]
  150. Wathes, D.C.; Abayasekara, D.R.; Aitken, R.J. Polyunsaturated fatty acids in male and female reproduction. Biol. Reprod. 2007, 77, 190–201. [Google Scholar] [CrossRef]
  151. Falsig, A.-M.L.; Gleerup, C.S.; Knudsen, U.B. The influence of omega-3 fatty acids on semen quality markers: A systematic PRISMA review. Andrology 2019, 7, 794–803. [Google Scholar] [CrossRef]
  152. Brown, S.A.; Brown, C.A.; Crowell, W.A.; Barsanti, J.A.; Kang, C.-W.; Allen, T.; Cowell, C.; Finco, D.R. Effects of dietary polyunsaturated fatty acid supplementation in early renal insufficiency in dogs. J. Lab. Clin. Med. 2000, 135, 275–286. [Google Scholar] [CrossRef]
  153. Brown, S.A.; Brown, C.A.; Crowell, W.A.; Barsanti, J.A.; Allen, T.; Cowell, C.; Finco, D.R. Beneficial effects of chronic administration of dietary ω-3 polyunsaturated fatty acids in dogs with renal insufficiency. J. Lab. Clin. Med. 1998, 131, 447–455. [Google Scholar] [CrossRef]
  154. Salem, N., Jr.; Litman, B.; Kim, H.-Y.; Gawrisch, K. Mechanisms of action of docosahexaenoic acid in the nervous system. Lipids 2001, 36, 945–959. [Google Scholar] [CrossRef]
  155. McNamara, R.K.; Carlson, S.E. Role of omega-3 fatty acids in brain development and function: Potential implications for the pathogenesis and prevention of psychopathology. Prostaglandins Leukot Essent Fat. Acids 2006, 75, 329–349. [Google Scholar] [CrossRef] [PubMed]
  156. Blanchard, T.; Eppe, J.; Mugnier, A.; Delfour, F.; Meynadier, A. Enhancing cognitive functions in aged dogs and cats: A systematic review of enriched diets and nutraceuticals. GeroScience 2025, 47, 2925–2947. [Google Scholar] [CrossRef] [PubMed]
  157. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  158. Fuller, R. Probiotics in man and animals. J. Appl. Bacteriol. 1989, 66, 365–378. [Google Scholar] [CrossRef]
  159. Kayser, E.; He, F.; Nixon, S.; Howard-Varona, A.; Lamelas, A.; Martinez-Blanch, J.; Chenoll, E.; Davenport, G.M.; de Godoy, M.R.C. Effects of supplementation of live and heat-treated Bifidobacterium animalis subspecies lactis CECT 8145 on glycemic and insulinemic response, fecal microbiota, systemic biomarkers of inflammation, and white blood cell gene expression of adult dogs. J. Anim. Sci. 2024, 102, skae291. [Google Scholar] [CrossRef]
  160. Bampidis, V.; Azimonti, G.; de Lourdes Bastos, M.; Christensen, H.; Dusemund, B.; Fa smon Durjava, M.; Kouba, M.; López-Alonso, M.; López Puente, S.; Marcon, F.; et al. Safety and efficacy of a feed additive consisting of Bacillus velezensis DSM 15544 (Calsporin ®) for piglets (suckling and weaned), pigs for fattening, sows in order to have benefit in piglets, ornamental fish, dogs and all avian species (Asahi Biocycle Co.). EFSA J. 2021, 19, 6903. [Google Scholar] [CrossRef]
  161. Yang, Q.; Wu, Z. Gut Probiotics and Health of Dogs and Cats: Benefits, Applications, and Underlying Mechanisms. Microorganisms 2023, 11, 2452. [Google Scholar] [CrossRef]
  162. Jugan, M.C.; Rudinsky, A.J.; Gordon, A.; Kramer, D.L.; Daniels, J.B.; Paliy, O.; Boyaka, P.; Gilor, C. Effects of oral Akkermansia muciniphila supplementation in healthy dogs following antimicrobial administration. Am. J. Vet. Res. 2018, 79, 884–892. [Google Scholar] [CrossRef]
  163. Xu, J.; Wen, C.; Song, G.; Lesaux, A.A.; Zhang, H.; Luo, Y. Effect of yeast probiotic Saccharomyces cerevisiae on the gut health of dogs undergoing rapid dietary transition. Front. Microbiol. 2025, 16, 1561660. [Google Scholar] [CrossRef] [PubMed]
  164. Marshall-Jones, Z.V.; Baillon, M.-L.A.; Croft, J.M.; Butterwick, R.F. Effects of Lactobacillus acidophilus DSM13241 as a probiotic in healthy adult cats. Am. J. Vet. Res. 2006, 67, 1005–1012. [Google Scholar] [CrossRef] [PubMed]
  165. De Filippis, F.; Esposito, A.; Ercolini, D. Outlook on next-generation probiotics from the human gut. Cell. Mol. Life Sci. 2022, 79, 76. [Google Scholar] [CrossRef] [PubMed]
  166. Chang, C.-J.; Lin, T.-L.; Tsai, Y.-L.; Wu, T.-R.; Lai, W.-F.; Lu, C.-C.; Lai, H.-C. Next generation probiotics in disease amelioration. J. Food Drug Anal. 2019, 27, 615–622. [Google Scholar] [CrossRef]
  167. Grześkowiak, Ł.; Endo, A.; Beasley, S.; Salminen, S. Microbiota and probiotics in canine and feline welfare. Anaerobe 2015, 34, 14–23. [Google Scholar] [CrossRef]
  168. Gorzelanna, Z.; Mamrot, A.; Będkowska, D.; Bubak, J.; Miszczak, M. Exploring the Potential of Novel Animal-Origin Probiotics as Key Players in One Health: Opportunities and Challenges. Int. J. Mol. Sci. 2025, 26, 5143. [Google Scholar] [CrossRef]
  169. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef]
  170. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
  171. Ravanal, M.C.; Contador, C.A.; Wong, W.-T.; Zhang, Q.; Roman-Benn, A.; Ah-Hen, K.S.; Ulloa, P.E.; Lam, H.-M. Prebiotics in animal nutrition: Harnessing agro-industrial waste for improved gut health and performance. Anim. Nutr. 2025, 21, 179–192. [Google Scholar] [CrossRef]
  172. Rose, L.; Rose, J.; Gosling, S.; Holmes, M. Efficacy of a Probiotic-Prebiotic Supplement on Incidence of Diarrhea in a Dog Shelter: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Vet. Int. Med. 2017, 31, 377–382. [Google Scholar] [CrossRef] [PubMed]
  173. Armian, A.M.; Pourjafar, H.; Gharamaleki, M.N. Study of the Effect of Synbiotic Diet on Haematological and Oxidative Indexes Changes in Male Dogs. Vet. Med. Sci. 2025, 11, e70290. [Google Scholar] [CrossRef] [PubMed]
  174. Fernandez-Pinteno, A.; Pilla, R.; Manteca, X.; Suchodolski, J.; Torre, C.; Salas-Mani, A. Age-associated changes in intestinal health biomarkers in dogs. Front. Vet. Sci. 2023, 10, 1213287. [Google Scholar] [CrossRef] [PubMed]
  175. Shah, H.; Trivedi, M.; Gurjar, T.; Sahoo, D.K.; Jergens, A.E.; Yadav, V.K.; Patel, A.; Pandya, P. Decoding the Gut Microbiome in Companion Animals: Impacts and Innovations. Microorganisms 2024, 12, 1831. [Google Scholar] [CrossRef]
  176. Beloshapka, A.N.; Wolff, A.K.; Swanson, K.S. Effects of feeding polydextrose on faecal characteristics, microbiota and fermentative end products in healthy adult dogs. Br. J. Nutr. 2012, 108, 638–644. [Google Scholar] [CrossRef]
  177. Perini, M.P.; Pedrinelli, V.; Marchi, P.H.; Henríquez, L.B.F.; Zafalon, R.V.A.; Vendramini, T.H.A.; Balieiro, J.C.d.C.; Brunetto, M.A. Potential Effects of Prebiotics on Gastrointestinal and Immunological Modulation in the Feeding of Healthy Dogs: A Review. Fermentation 2023, 9, 693. [Google Scholar] [CrossRef]
  178. Bosch, G.; Verbrugghe, A.; Hesta, M.; Holst, J.J.; van der Poel, A.F.; Janssens, G.P.; Hendriks, W.H. The effects of dietary fibre type on satiety-related hormones and voluntary food intake in dogs. Br. J. Nutr. 2009, 102, 318–325. [Google Scholar] [CrossRef]
  179. Alexander, C.; Cross, T.L.; Devendran, S.; Neumer, F.; Theis, S.; Ridlon, J.M.; Suchodolski, J.S.; de Godoy, M.R.C.; Swanson, K.S. Effects of prebiotic inulin-type fructans on blood metabolite and hormone concentrations and faecal microbiota and metabolites in overweight dogs. Br. J. Nutr. 2018, 120, 711–720. [Google Scholar] [CrossRef]
  180. Respondek, F.; Swanson, K.S.; Belsito, K.R.; Vester, B.M.; Wagner, A.; Istasse, L.; Diez, M. Short-Chain Fructooligosaccharides Influence Insulin Sensitivity and Gene Expression of Fat Tissue in Obese Dogs12. J. Nutr. 2008, 138, 1712–1718. [Google Scholar] [CrossRef]
  181. Pilla, R.; Suchodolski, J.S. The Gut Microbiome of Dogs and Cats, and the Influence of Diet. Vet. Clin. N. Am. Small Anim. Pract. 2021, 51, 605–621. [Google Scholar] [CrossRef]
  182. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef] [PubMed]
  183. Li, H.Y.; Zhou, D.D.; Gan, R.Y.; Huang, S.Y.; Zhao, C.N.; Shang, A.; Xu, X.Y.; Li, H.B. Effects and Mechanisms of Probiotics, Prebiotics, Synbiotics, and Postbiotics on Metabolic Diseases Targeting Gut Microbiota: A Narrative Review. Nutrients 2021, 13, 3211. [Google Scholar] [CrossRef] [PubMed]
  184. Yan, F.; Polk, D.B. Probiotics and Probiotic-Derived Functional Factors—Mechanistic Insights Into Applications for Intestinal Homeostasis. Front. Immunol. 2020, 11, 1428. [Google Scholar] [CrossRef] [PubMed]
  185. Bastos, T.S.; Souza, C.M.M.; Legendre, H.; Richard, N.; Pilla, R.; Suchodolski, J.S.; de Oliveira, S.G.; Lesaux, A.A.; Félix, A.P. Effect of Yeast Saccharomyces cerevisiae as a Probiotic on Diet Digestibility, Fermentative Metabolites, and Composition and Functional Potential of the Fecal Microbiota of Dogs Submitted to an Abrupt Dietary Change. Microorganisms 2023, 11, 506. [Google Scholar] [CrossRef]
  186. Bastos, T.S.; de Lima, D.C.; Souza, C.M.M.; Maiorka, A.; de Oliveira, S.G.; Bittencourt, L.C.; Félix, A.P. Bacillus subtilis and Bacillus licheniformis reduce faecal protein catabolites concentration and odour in dogs. BMC Vet. Res. 2020, 16, 116. [Google Scholar] [CrossRef]
  187. Sun, H.Y.; Kim, K.P.; Bae, C.H.; Choi, A.J.; Paik, H.D.; Kim, I.H. Evaluation of Weissella Cibaria JW15 Probiotic Derived from Fermented Korean Vegetable Product Supplementation in Diet on Performance Characteristics in Adult Beagle Dog. Animals 2019, 9, 581. [Google Scholar] [CrossRef]
  188. Fusi, E.; Rizzi, R.; Polli, M.; Cannas, S.; Giardini, A.; Bruni, N.; Marelli, S.P. Effects of Lactobacillus acidophilus D2/CSL (CECT 4529) supplementation on healthy cat performance. Vet. Rec. Open 2019, 6, e000368. [Google Scholar] [CrossRef]
  189. Li, Y.; Ali, I.; Lei, Z.; Li, Y.; Yang, M.; Yang, C.; Li, L. Effect of a Multistrain Probiotic on Feline Gut Health through the Fecal Microbiota and Its Metabolite SCFAs. Metabolites 2023, 13, 228. [Google Scholar] [CrossRef]
  190. Rossi, G.; Gioacchini, G.; Pengo, G.; Suchodolski, J.S.; Jergens, A.E.; Allenspach, K.; Gavazza, A.; Scarpona, S.; Berardi, S.; Galosi, L.; et al. Enterocolic increase of cannabinoid receptor type 1 and type 2 and clinical improvement after probiotic administration in dogs with chronic signs of colonic dysmotility without mucosal inflammatory changes. Neurogastroenterol. Motil. 2020, 32, e13717. [Google Scholar] [CrossRef]
  191. Xu, H.; Zhao, F.; Hou, Q.; Huang, W.; Liu, Y.; Zhang, H.; Sun, Z. Metagenomic analysis revealed beneficial effects of probiotics in improving the composition and function of the gut microbiota in dogs with diarrhoea. Food Funct. 2019, 10, 2618–2629. [Google Scholar] [CrossRef]
  192. Ziese, A.-L.; Suchodolski, J.S.; Hartmann, K.; Busch, K.; Anderson, A.; Sarwar, F.; Sindern, N.; Unterer, S. Effect of probiotic treatment on the clinical course, intestinal microbiome, and toxigenic Clostridium perfringens in dogs with acute hemorrhagic diarrhea. PLoS ONE 2018, 13, e0204691. [Google Scholar] [CrossRef]
  193. Zha, M.; Zhu, S.; Chen, Y. Probiotics and Cat Health: A Review of Progress and Prospects. Microorganisms 2024, 12, 1080. [Google Scholar] [CrossRef] [PubMed]
  194. Tsai, C.-W.; Huang, H.-W.; Lee, Y.-J.; Chen, M.-J. Investigating the Efficacy of Kidney-Protective Lactobacillus Mixture-Containing Pet Treats in Feline Chronic Kidney Disease and Its Possible Mechanism. Animals 2024, 14, 630. [Google Scholar] [CrossRef] [PubMed]
  195. Ansari, F.; Neshat, M.; Pourjafar, H.; Jafari, S.M.; Samakkhah, S.A.; Mirzakhani, E. The role of probiotics and prebiotics in modulating of the gut-brain axis. Front. Nutr. 2023, 10, 1173660. [Google Scholar] [CrossRef] [PubMed]
  196. Mondo, E.; Barone, M.; Soverini, M.; D’Amico, F.; Cocchi, M.; Petrulli, C.; Mattioli, M.; Marliani, G.; Candela, M.; Accorsi, P.A. Gut microbiome structure and adrenocortical activity in dogs with aggressive and phobic behavioral disorders. Heliyon 2020, 6, e03311. [Google Scholar] [CrossRef]
  197. Kirchoff, N.S.; Udell, M.A.R.; Sharpton, T.J. The gut microbiome correlates with conspecific aggression in a small population of rescued dogs (Canis familiaris). PeerJ 2019, 7, e6103. [Google Scholar] [CrossRef]
  198. Pellowe, S.D.; Zhang, A.; Bignell, D.R.D.; Pena-Castillo, L.; Walsh, C.J. Gut microbiota composition is related to anxiety and aggression scores in companion dogs. Sci. Rep. 2025, 15, 24336. [Google Scholar] [CrossRef]
  199. Yeh, Y.-M.; Lye, X.-Y.; Lin, H.-Y.; Wong, J.-Y.; Wu, C.-C.; Huang, C.-L.; Tsai, Y.-C.; Wang, L.-C. Effects of Lactiplantibacillus plantarum PS128 on alleviating canine aggression and separation anxiety. Appl. Anim. Behav. Sci. 2022, 247, 105569. [Google Scholar] [CrossRef]
  200. Bijaoui, E.M.M.; Zimmerman, N.P. Efficacy of a Novel Lactiplantibacillus plantarum Strain (LP815TM) in Reducing Canine Aggression and Anxiety: A Randomized Placebo-Controlled Trial with Qualitative and Quantitative Assessment. Animals 2025, 15, 2280. [Google Scholar] [CrossRef]
  201. Markowiak, P.; Śliżewska, K. The role of probiotics, prebiotics and synbiotics in animal nutrition. Gut Pathog. 2018, 10, 21. [Google Scholar] [CrossRef]
  202. Liu, X.; Zhao, H.; Wong, A. Accounting for the health risk of probiotics. Heliyon 2024, 10, e27908. [Google Scholar] [CrossRef]
  203. Kerek, A.; Szabó, E.; Szabó, Á.; Papp, M.; Bányai, K.; Kardos, G.; Kaszab, E.; Bali, K.; Jerzsele, Á. Investigating antimicrobial resistance genes in probiotic products for companion animals. Front. Vet. Sci. 2024, 11, 1464351. [Google Scholar] [CrossRef]
  204. Lee, D.; Goh, T.W.; Kang, M.G.; Choi, H.J.; Yeo, S.Y.; Yang, J.; Huh, C.S.; Kim, Y.Y.; Kim, Y. Perspectives and advances in probiotics and the gut microbiome in companion animals. J. Anim. Sci. Technol. 2022, 64, 197–217. [Google Scholar] [CrossRef]
  205. Romero, B.; Susperregui, J.; Sahagún, A.M.; Diez, M.J.; Fernández, N.; García, J.J.; López, C.; Sierra, M.; Díez, R. Use of medicinal plants by veterinary practitioners in Spain: A cross-sectional survey. Front. Vet. Sci. 2022, 9, 1060738. [Google Scholar] [CrossRef]
  206. Kumar, A.; P, N.; Kumar, M.; Jose, A.; Tomer, V.; Oz, E.; Proestos, C.; Zeng, M.; Elobeid, T.; K, S.; et al. Major Phytochemicals: Recent Advances in Health Benefits and Extraction Method. Molecules 2023, 28, 887. [Google Scholar] [CrossRef] [PubMed]
  207. Ivanova, S.; Sukhikh, S.; Popov, A.; Shishko, O.; Nikonov, I.; Kapitonova, E.; Krol, O.; Larina, V.; Noskova, S.; Babich, O. Medicinal plants: A source of phytobiotics for the feed additives. J. Agric. Food Res. 2024, 16, 101172. [Google Scholar] [CrossRef]
  208. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  209. Waheed Janabi, A.H.; Kamboh, A.A.; Saeed, M.; Xiaoyu, L.; BiBi, J.; Majeed, F.; Naveed, M.; Mughal, M.J.; Korejo, N.A.; Kamboh, R.; et al. Flavonoid-rich foods (FRF): A promising nutraceutical approach against lifespan-shortening diseases. Iran. J. Basic Med. Sci. 2020, 23, 140–153. [Google Scholar] [CrossRef]
  210. Li, A.-N.; Li, S.; Zhang, Y.-J.; Xu, X.-R.; Chen, Y.-M.; Li, H.-B. Resources and Biological Activities of Natural Polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef]
  211. Green, A.S.; Fascetti, A.J. Meeting the Vitamin A Requirement: The Efficacy and Importance of beta-Carotene in Animal Species. Sci. World J. 2016, 2016, 7393620. [Google Scholar] [CrossRef]
  212. Salehi, B.; Quispe, C.; Sharifi-Rad, J.; Cruz-Martins, N.; Nigam, M.; Mishra, A.P.; Konovalov, D.A.; Orobinskaya, V.; Abu-Reidah, I.M.; Zam, W.; et al. Phytosterols: From Preclinical Evidence to Potential Clinical Applications. Front. Pharmacol. 2020, 11, 599959. [Google Scholar] [CrossRef]
  213. Borin-Crivellenti, S.; Crivellenti, L.Z.; de Oliveira, F.R.; Costa, P.B.; Alvarenga, A.W.O.; Rezende, L.R.; Gouvêa, F.N.; Assef, N.D.; Branco, L.d.O. Effect of phytosterols on reducing low-density lipoprotein cholesterol in dogs. Domest. Anim. Endocrinol. 2021, 76, 106610. [Google Scholar] [CrossRef] [PubMed]
  214. Reichling, J.; Schmokel, H.; Fitzi, J.; Bucher, S.; Saller, R. Dietary support with Boswellia resin in canine inflammatory joint and spinal disease. Schweiz. Arch. Tierheilkd. 2004, 146, 71–79. [Google Scholar] [CrossRef] [PubMed]
  215. Metwaly, A.M.; Lianlian, Z.; Luqi, H.; Deqiang, D. Black Ginseng and Its Saponins: Preparation, Phytochemistry and Pharmacological Effects. Molecules 2019, 24, 1856. [Google Scholar] [CrossRef] [PubMed]
  216. Qu, Q.; Zhang, W.; Xuan, Z.; Chen, R.; Ma, Y.; Huang, Y.; Hu, Y.; Lin, Y.; Liu, M.; Lv, W.; et al. Evaluation of Anti-Inflammatory Effects of Six Ginsenosides and Rg1 Regulation of Macrophage Polarization and Metabolites to Alleviate Colitis. Antioxidants 2025, 14, 283. [Google Scholar] [CrossRef]
  217. Siddiqui, M.Z. Boswellia serrata, a potential antiinflammatory agent: An overview. Indian J. Pharm. Sci. 2011, 73, 255–261. [Google Scholar]
  218. Umar, S.; Umar, K.; Sarwar, A.H.M.G.; Khan, A.; Ahmad, N.; Ahmad, S.; Katiyar, C.K.; Husain, S.A.; Khan, H.A. Boswellia serrata extract attenuates inflammatory mediators and oxidative stress in collagen induced arthritis. Phytomedicine 2014, 21, 847–856. [Google Scholar] [CrossRef]
  219. Lee, K.W.; Yamato, O.; Tajima, M.; Kuraoka, M.; Omae, S.; Maede, Y. Hematologic changes associated with the appearance of eccentrocytes after intragastric administration of garlic extract to dogs. Am. J. Vet. Res. 2000, 61, 1446–1450. [Google Scholar] [CrossRef]
  220. Quintavalla, F. Phytotherapeutic Approaches in Canine Pediatrics. Vet. Sci. 2024, 11, 133. [Google Scholar] [CrossRef]
  221. Burns, K. Assessing pet supplements American Veterinary Medical Association. 2017. Available online: https://www.avma.org/javma-news/2017-01-15/assessing-pet-supplements#:~:text=The%20supplements%20that%20hold%20promise,been%20shown%20to%20be%20questionable (accessed on 18 June 2025).
  222. Youness, R.A.; Dawoud, A.; ElTahtawy, O.; Farag, M.A. Fat-soluble vitamins: Updated review of their role and orchestration in human nutrition throughout life cycle with sex differences. Nutr. Metab. 2022, 19, 60. [Google Scholar] [CrossRef]
  223. Lykstad, J.; Sharma, S. Biochemistry, Water Soluble Vitamins. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  224. Shastak, Y.; Pelletier, W. Pet Wellness and Vitamin A: A Narrative Overview. Animals 2024, 14, 1000. [Google Scholar] [CrossRef] [PubMed]
  225. Kritikos, G.; Parr, J.M.; Verbrugghe, A. The Role of Thiamine and Effects of Deficiency in Dogs and Cats. Vet. Sci. 2017, 4, 59. [Google Scholar] [CrossRef]
  226. EFSA Panel on Additives and Products or Substances used in Animal Feed (EFSA FEEDAP Panel); Rychen, G.; Aquilina, G.; Azimonti, G.; Bampidis, V.; Bastos, M.d.L.; Bories, G.; Chesson, A.; Cocconcelli, P.S.; Flachowsky, G.; et al. Safety and efficacy of vitamin B2 (riboflavin) produced by Ashbya gossypii DSM 23096 for all animal species based on a dossier submitted by BASF SE. EFSA J. 2018, 16, e05337. [Google Scholar] [CrossRef]
  227. Chu, V.; Fascetti, A.J.; Larsen, J.A.; Montano, M.; Giulivi, C. Factors influencing vitamin B6 status in domestic cats: Age, disease, and body condition score. Sci. Rep. 2024, 14, 2037. [Google Scholar] [CrossRef]
  228. Gordon, D.S.; Rudinsky, A.J.; Guillaumin, J.; Parker, V.J.; Creighton, K.J. Vitamin C in Health and Disease: A Companion Animal Focus. Top. Companion Anim. Med. 2020, 39, 100432. [Google Scholar] [CrossRef]
  229. Clarke, K.E.; Hurst, E.A.; Mellanby, R.J. Vitamin D metabolism and disorders in dogs and cats. J. Small Anim. Pract. 2021, 62, 935–947. [Google Scholar] [CrossRef]
  230. Mellanby, R.J. Beyond the skeleton: The role of vitamin D in companion animal health. J. Small Anim. Pract. 2016, 57, 175–180. [Google Scholar] [CrossRef]
  231. Jewell, D.E.; Motsinger, L.A.; Paetau-Robinson, I. Effect of dietary antioxidants on free radical damage in dogs and cats. J. Anim. Sci. 2024, 102, skae153. [Google Scholar] [CrossRef]
  232. Kohn, B.; Weingart, C.; Giger, U. Haemorrhage in seven cats with suspected anticoagulant rodenticide intoxication. J. Feline Med. Surg. 2003, 5, 295–304. [Google Scholar] [CrossRef]
  233. Mooney, E.; Agostini, G.; Griebsch, C.; Hickey, M. Intravenous vitamin K1 normalises prothrombin time in 1 hour in dogs with anticoagulant rodenticide toxicosis. Aust. Vet. J. 2020, 98, 225–231. [Google Scholar] [CrossRef]
  234. Sadler, R.A.; Shoveller, A.K.; Shandilya, U.K.; Charchoglyan, A.; Wagter-Lesperance, L.; Bridle, B.W.; Mallard, B.A.; Karrow, N.A. Beyond the Coagulation Cascade: Vitamin K and Its Multifaceted Impact on Human and Domesticated Animal Health. Curr. Issues Mol. Biol. 2024, 46, 7001–7031. [Google Scholar] [CrossRef]
  235. Li, P.; Wu, G. Characteristics of Nutrition and Metabolism in Dogs and Cats. Adv. Exp. Med. Biol. 2024, 1446, 55–98. [Google Scholar] [CrossRef]
  236. Irungbam, K.; Chavhan, S.; Kulkarni, S.; Sonphule, A.; Naik, L.; Hanah, S. Hypervitaminosis or Vitamin Poisoning in Animals. North-East Vet. 2013, 13, 3–6+8. [Google Scholar]
  237. Crossley, V.J.; Bovens, C.P.; Pineda, C.; Hibbert, A.; Finch, N.C. Vitamin D toxicity of dietary origin in cats fed a natural complementary kitten food. JFMS Open Rep. 2017, 3, 2055116917743613. [Google Scholar] [CrossRef] [PubMed]
  238. Mellanby, R.J.; Mee, A.P.; Berry, J.L.; Herrtage, M.E. Hypercalcaemia in two dogs caused by excessive dietary supplementation of vitamin D. J. Small Anim. Pract. 2005, 46, 334–338. [Google Scholar] [CrossRef] [PubMed]
  239. Siddiqui, K.; Bawazeer, N.; Scaria Joy, S. Variation in Macro and Trace Elements in Progression of Type 2 Diabetes. Sci. World J. 2014, 2014, 461591. [Google Scholar] [CrossRef]
  240. Stepanova, M.V.; Sotnikova, L.F.; Zaitsev, S.Y. Relationships between the Content of Micro- and Macroelements in Animal Samples and Diseases of Different Etiologies. Animals 2023, 13, 852. [Google Scholar] [CrossRef]
  241. Pajarillo, E.A.B.; Lee, E.; Kang, D.-K. Trace metals and animal health: Interplay of the gut microbiota with iron, manganese, zinc, and copper. Anim. Nutr. 2021, 7, 750–761. [Google Scholar] [CrossRef]
  242. Razzaque, M.S.; Wimalawansa, S.J. Minerals and Human Health: From Deficiency to Toxicity. Nutrients 2025, 17, 454. [Google Scholar] [CrossRef]
  243. Mondola, P.; Damiano, S.; Sasso, A.; Santillo, M. The Cu, Zn Superoxide Dismutase: Not Only a Dismutase Enzyme. Front. Physiol. 2016, 7, 594. [Google Scholar] [CrossRef]
  244. Pereira, A.M.; Maia, M.R.G.; Fonseca, A.J.M.; Cabrita, A.R.J. Zinc in Dog Nutrition, Health and Disease: A Review. Animals 2021, 11, 978. [Google Scholar] [CrossRef]
  245. Colombini, S. Canine Zinc-Responsive Dermatosis. Vet. Clin. N. Am. Small Anim. Pract. 1999, 29, 1373–1383. [Google Scholar] [CrossRef]
  246. van den Broek, A.H.M.; Thoday, K.L. Skin disease in dogs associated with zinc deficiency: A report of five cases. J. Small Anim. Pract. 1986, 27, 313–323. [Google Scholar] [CrossRef]
  247. White, S.D.; Bourdeau, P.; Rosychuk, R.A.W.; Cohen, B.; Bonenberger, T.; Fieseler, K.V.; Ihrke, P.; Chapman, P.L.; Schultheiss, P.; Zur, G.; et al. Zinc-responsive dermatosis in dogs: 41 cases and literature review. Vet. Dermatol. 2001, 12, 101–109. [Google Scholar] [CrossRef] [PubMed]
  248. Soltanian, A.; Khoshnegah, J.; Heidarpour, M. Comparison of serum trace elements and antioxidant levels in terrier dogs with or without behavior problems. Appl. Anim. Behav. Sci. 2016, 180, 87–92. [Google Scholar] [CrossRef]
  249. Yu, J.; Jenkins, E.; Podadera, J.M.; Proschogo, N.; Chan, R.; Boland, L. Zinc toxicosis in a cat associated with ingestion of a metal screw nut. J. Feline Med. Surg. Open Rep. 2022, 8, 20551169221136464. [Google Scholar] [CrossRef]
  250. Vitale, S.; Hague, D.W.; Foss, K.; de Godoy, M.C.; Selmic, L.E. Comparison of Serum Trace Nutrient Concentrations in Epileptics Compared to Healthy Dogs. Front. Vet. Sci. 2019, 6, 467. [Google Scholar] [CrossRef]
  251. Stockman, J.; Villaverde, C.; Corbee, R.J. Calcium, Phosphorus, and Vitamin D in Dogs and Cats: Beyond the Bones. Vet. Clin. N. Am. Small Anim. Pract. 2021, 51, 623–634. [Google Scholar] [CrossRef]
  252. Bailey, L.E.; Ong, S.D.; Queen, G.M. Calcium movement during contraction in the cat heart. J. Mol. Cell. Cardiol. 1972, 4, 121–138. [Google Scholar] [CrossRef]
  253. Laflamme, D.; Backus, R.; Brown, S.; Butterwick, R.; Czarnecki-Maulden, G.; Elliott, J.; Fascetti, A.; Polzin, D. A review of phosphorus homeostasis and the impact of different types and amounts of dietary phosphate on metabolism and renal health in cats. J. Vet. Int. Med. 2020, 34, 2187–2196. [Google Scholar] [CrossRef]
  254. Anand, A.; Aoyagi, H. Understudied Hyperphosphatemia (Chronic Kidney Disease) Treatment Targets and New Biological Approaches. Medicina 2023, 59, 959. [Google Scholar] [CrossRef]
  255. Groman, R.P. Acute management of calcium disorders. Top. Companion. Anim. Med. 2012, 27, 167–171. [Google Scholar] [CrossRef]
  256. Holowaychuk, M.K.; Hansen, B.D.; DeFrancesco, T.C.; Marks, S.L. Ionized Hypocalcemia in Critically Ill Dogs. J. Vet. Int. Med. 2009, 23, 509–513. [Google Scholar] [CrossRef] [PubMed]
  257. Holowaychuk, M.K. Hypocalcemia of critical illness in dogs and cats. Vet. Clin. N. Am. Small Anim. Pract. 2013, 43, 1299–1317. [Google Scholar] [CrossRef] [PubMed]
  258. Dobenecker, B.; Kienzle, E.; Siedler, S. The Source Matters–Effects of High Phosphate Intake from Eight Different Sources in Dogs. Animals 2021, 11, 3456. [Google Scholar] [CrossRef] [PubMed]
  259. Blanca, P.-M.; María Luisa, F.-R.; Guadalupe, M.; Fátima, C.-L. Oxidative Stress in Canine Diseases: A Comprehensive Review. Antioxidants 2024, 13, 1396. [Google Scholar] [CrossRef]
  260. Gu, X.; Gao, C.-q. New horizons for selenium in animal nutrition and functional foods. Anim. Nutr. 2022, 11, 80–86. [Google Scholar] [CrossRef]
  261. Zentrichová, V.; Pechová, A.; Kovaříková, S. Selenium and Dogs: A Systematic Review. Animals 2021, 11, 418. [Google Scholar] [CrossRef]
  262. Liu, Y.; Li, W.; Guo, M.; Li, C.; Qiu, C. Protective Role of Selenium Compounds on the Proliferation, Apoptosis, and Angiogenesis of a Canine Breast Cancer Cell Line. Biol. Trace Elem. Res. 2016, 169, 86–93. [Google Scholar] [CrossRef]
  263. Fico, M.E.; Poirier, K.A.; Watrach, A.M.; Watrach, M.A.; Milner, J.A. Differential effects of selenium on normal and neoplastic canine mammary cells. Cancer Res. 1986, 46, 3384–3388. [Google Scholar]
  264. Chiang, E.C.; Bostwick, D.G.; Waters, D.J. Homeostatic housecleaning effect of selenium: Evidence that noncytotoxic oxidant-induced damage sensitizes prostate cancer cells to organic selenium-triggered apoptosis. Biofactors 2013, 39, 575–588. [Google Scholar] [CrossRef]
  265. Soetan, K.; Olaiya, C.; Oyewole, O. The importance of mineral elements for humans, domestic animals and plants: A review. Afr. J. Food Sci. 2009, 4, 200–222. [Google Scholar]
  266. Adam, F.; Elliott, J.; Dandrieux, J.; Blackwood, L. Poisoning: Zinc toxicity in two dogs associated with the ingestion of identification tags. Vet. Rec. 2011, 168, 84–85. [Google Scholar] [CrossRef] [PubMed]
  267. Brutlag, A.G.; Flint, C.T.C.; Puschner, B. Iron Intoxication in a Dog Consequent to the Ingestion of Oxygen Absorber Sachets in Pet Treat Packaging. J. Med. Toxicol. 2012, 8, 76–79. [Google Scholar] [CrossRef] [PubMed]
  268. Wu, G.; Fang, Y.Z.; Yang, S.; Lupton, J.R.; Turner, N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004, 134, 489–492. [Google Scholar] [CrossRef]
  269. Pizzorno, J. Glutathione! Integr. Med. 2014, 13, 8–12. [Google Scholar]
  270. Mari, M.; Morales, A.; Colell, A.; Garcia-Ruiz, C.; Fernandez-Checa, J.C. Mitochondrial glutathione, a key survival antioxidant. Antioxid. Redox. Signal. 2009, 11, 2685–2700. [Google Scholar] [CrossRef]
  271. Biswas, S.K.; Rahman, I. Environmental toxicity, redox signaling and lung inflammation: The role of glutathione. Mol. Asp. Med. 2009, 30, 60–76. [Google Scholar] [CrossRef]
  272. Pizzorno, J. Is Mercury Toxicity an Epidemic? (Part II). Integr. Med. 2009, 8, 8–12. [Google Scholar]
  273. Averill-Bates, D.A. The antioxidant glutathione. Vitam. Horm. 2023, 121, 109–141. [Google Scholar] [CrossRef]
  274. Fiser, B.; Jojart, B.; Csizmadia, I.G.; Viskolcz, B. Glutathione--hydroxyl radical interaction: A theoretical study on radical recognition process. PLoS ONE 2013, 8, e73652. [Google Scholar] [CrossRef]
  275. Haddad, J.J.; Harb, H.L. L-gamma-Glutamyl-L-cysteinyl-glycine (glutathione; GSH) and GSH-related enzymes in the regulation of pro- and anti-inflammatory cytokines: A signaling transcriptional scenario for redox(y) immunologic sensor(s)? Mol. Immunol. 2005, 42, 987–1014. [Google Scholar] [CrossRef] [PubMed]
  276. Michałek, M.; Tabiś, A.; Pasławska, U.; Noszczyk-Nowak, A. Antioxidant defence and oxidative stress markers in cats with asymptomatic and symptomatic hypertrophic cardiomyopathy: A pilot study. BMC Vet. Res. 2020, 16, 26. [Google Scholar] [CrossRef] [PubMed]
  277. Ruparell, A.; Alexander, J.E.; Eyre, R.; Carvell-Miller, L.; Leung, Y.B.; Evans, S.J.M.; Holcombe, L.J.; Heer, M.; Watson, P. Glycine supplementation can partially restore oxidative stress-associated glutathione deficiency in ageing cats. Br. J. Nutr. 2024, 131, 1947–1961. [Google Scholar] [CrossRef]
  278. Jewell, D.E.; Toll, P.W.; Wedekind, K.J.; Zicker, S.C. Effect of increasing dietary antioxidants on concentrations of vitamin E and total alkenals in serum of dogs and cats. Vet. Ther. 2000, 1, 264–272. [Google Scholar]
  279. Viviano, K.R.; Lavergne, S.N.; Goodman, L.; Vanderwielen, B.; Grundahl, L.; Padilla, M.; Trepanier, L.A. Glutathione, cysteine, and ascorbate concentrations in clinically ill dogs and cats. J. Vet. Int. Med. 2009, 23, 250–257. [Google Scholar] [CrossRef]
  280. Center, S.A.; Warner, K.L.; Erb, H.N. Liver glutathione concentrations in dogs and cats with naturally occurring liver disease. Am. J. Vet. Res. 2002, 63, 1187–1197. [Google Scholar] [CrossRef]
  281. Burgunder, J.M.; Lauterburg, B.H. Decreased production of glutathione in patients with cirrhosis. Eur. J. Clin. Investig. 1987, 17, 408–414. [Google Scholar] [CrossRef]
  282. Lu, S.C. Dysregulation of glutathione synthesis in liver disease. Liver Res. 2020, 4, 64–73. [Google Scholar] [CrossRef]
  283. Weschawalit, S.; Thongthip, S.; Phutrakool, P.; Asawanonda, P. Glutathione and its antiaging and antimelanogenic effects. Clin. Cosmet. Investig. Dermatol. 2017, 10, 147–153. [Google Scholar] [CrossRef]
  284. Sinha, R.; Sinha, I.; Calcagnotto, A.; Trushin, N.; Haley, J.S.; Schell, T.D.; Richie, J.P., Jr. Oral supplementation with liposomal glutathione elevates body stores of glutathione and markers of immune function. Eur. J. Clin. Nutr. 2018, 72, 105–111. [Google Scholar] [CrossRef]
  285. Honda, Y.; Kessoku, T.; Sumida, Y.; Kobayashi, T.; Kato, T.; Ogawa, Y.; Tomeno, W.; Imajo, K.; Fujita, K.; Yoneda, M.; et al. Efficacy of glutathione for the treatment of nonalcoholic fatty liver disease: An open-label, single-arm, multicenter, pilot study. BMC Gastroenterol. 2017, 17, 96. [Google Scholar] [CrossRef]
  286. Vulcano, L.A.; Confalonieri, O.; Franci, R.; Tapia, M.O.; Soraci, A.L. Efficacy of free glutathione and niosomal glutathione in the treatment of acetaminophen-induced hepatotoxicity in cats. Open Vet. J. 2013, 3, 56–63. [Google Scholar] [CrossRef] [PubMed]
  287. Martello, E.; Perondi, F.; Bisanzio, D.; Lippi, I.; Meineri, G.; Gabriele, V. Antioxidant Effect of a Dietary Supplement Containing Fermentative S-Acetyl-Glutathione and Silybin in Dogs with Liver Disease. Vet. Sci. 2023, 10, 131. [Google Scholar] [CrossRef] [PubMed]
  288. Fernández-Martín, S.; González-Cantalapiedra, A.; Muñoz, F.; García-González, M.; Permuy, M.; López-Peña, M. Glucosamine and Chondroitin Sulfate: Is There Any Scientific Evidence for Their Effectiveness as Disease-Modifying Drugs in Knee Osteoarthritis Preclinical Studies?—A Systematic Review from 2000 to 2021. Animals 2021, 11, 1608. [Google Scholar] [CrossRef] [PubMed]
  289. Brito, R.; Costa, D.; Dias, C.; Cruz, P.; Barros, P. Chondroitin Sulfate Supplements for Osteoarthritis: A Critical Review. Cureus 2023, 15, e40192. [Google Scholar] [CrossRef]
  290. Rajesh, A.; Sajeev, D.; Kumaar, R.N.; Rangasamy, J.; Nair, S.C. Chondroitin sulfate: From bioactive molecule to versatile drug delivery system for advancing regenerative medicine. Int. J. Biol. Macromol. 2025, 311, 143746. [Google Scholar] [CrossRef]
  291. Kelso, J.M. Potential food allergens in medications. J. Allergy Clin. Immunol. 2014, 133, 1509–1518. [Google Scholar] [CrossRef]
  292. Henrotin, Y.; Mobasheri, A.; Marty, M. Is there any scientific evidence for the use of glucosamine in the management of human osteoarthritis? Arthritis Res. Ther. 2012, 14, 201. [Google Scholar] [CrossRef]
  293. Lippiello, L.; Woodward, J.; Karpman, R.; Hammad, T.A. In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin. Orthop. Relat. Res. 2000, 381, 229–240. [Google Scholar] [CrossRef]
  294. Sen, R.; Hurley, J.A. Osteoarthritis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  295. Anderson, K.L.; O’Neill, D.G.; Brodbelt, D.C.; Church, D.B.; Meeson, R.L.; Sargan, D.; Summers, J.F.; Zulch, H.; Collins, L.M. Prevalence, duration and risk factors for appendicular osteoarthritis in a UK dog population under primary veterinary care. Sci. Rep. 2018, 8, 5641. [Google Scholar] [CrossRef]
  296. Slingerland, L.I.; Hazewinkel, H.A.W.; Meij, B.P.; Picavet, P.; Voorhout, G. Cross-sectional study of the prevalence and clinical features of osteoarthritis in 100 cats. Vet. J. 2011, 187, 304–309. [Google Scholar] [CrossRef]
  297. Rychel, J.K. Diagnosis and treatment of osteoarthritis. Top. Companion. Anim. Med. 2010, 25, 20–25. [Google Scholar] [CrossRef] [PubMed]
  298. Wood, M.J.; Miller, R.E.; Malfait, A.M. The Genesis of Pain in Osteoarthritis: Inflammation as a Mediator of Osteoarthritis Pain. Clin. Geriatr. Med. 2022, 38, 221–238. [Google Scholar] [CrossRef] [PubMed]
  299. Epstein, M.E.; Rodanm, I.; Griffenhagen, G.; Kadrlik, J.; Petty, M.C.; Robertson, S.A.; Simpson, W. 2015 AAHA/AAFP pain management guidelines for dogs and cats. J. Feline Med. Surg. 2015, 17, 251–272. [Google Scholar] [CrossRef] [PubMed]
  300. Vasiliadis, H.S.; Tsikopoulos, K. Glucosamine and chondroitin for the treatment of osteoarthritis. World J. Orthop. 2017, 8, 1–11. [Google Scholar] [CrossRef]
  301. Bai, H.; Liu, T.; Wang, H.; Li, Y.; Wang, Z. Chondroitin sulfate alleviated lipopolysaccharide-induced arthritis in feline and canine articular chondrocytes through regulation of neurotrophic signaling pathways and apoptosis. Tissue Cell 2024, 91, 102642. [Google Scholar] [CrossRef]
  302. Silva, F.S.; Yoshinari, N.H.; Castro, R.R.; Girão, V.C.C.; Pompeu, M.M.L.; de Andrade Feitosa, J.P.; Rocha, F.A.C. Combined glucosamine and chondroitin sulfate provides functional and structural benefit in the anterior cruciate ligament transection model. Clin. Rheumatol. 2009, 28, 109–117. [Google Scholar] [CrossRef]
  303. Terencio, M.C.; Ferrándiz, M.L.; Carceller, M.C.; Ruhí, R.; Dalmau, P.; Vergés, J.; Montell, E.; Torrent, A.; Alcaraz, M.J. Chondroprotective effects of the combination chondroitin sulfate-glucosamine in a model of osteoarthritis induced by anterior cruciate ligament transection in ovariectomised rats. Biomed. Pharmacother. 2016, 79, 120–128. [Google Scholar] [CrossRef]
  304. McCarthy, G.; O’Donovan, J.; Jones, B.; McAllister, H.; Seed, M.; Mooney, C. Randomised double-blind, positive-controlled trial to assess the efficacy of glucosamine/chondroitin sulfate for the treatment of dogs with osteoarthritis. Vet. J. 2007, 174, 54–61. [Google Scholar] [CrossRef]
  305. Kampa, N.; Kaenkangploo, D.; Jitpean, S.; Srithunyarat, T.; Seesupa, S.; Hoisang, S.; Yongvanit, K.; Kamlangchai, P.; Tuchpramuk, P.; Lascelles, B.D.X. Study of the effectiveness of glucosamine and chondroitin sulfate, marine based fatty acid compounds (PCSO-524 and EAB-277), and carprofen for the treatment of dogs with hip osteoarthritis: A prospective, block-randomized, double-blinded, placebo-controlled clinical trial. Front. Vet. Sci. 2023, 10, 1033188. [Google Scholar] [CrossRef]
  306. Moreau, M.; Dupuis, J.; Bonneau, N.H.; Desnoyers, M. Clinical evaluation of a nutraceutical, carprofen and meloxicam for the treatment of dogs with osteoarthritis. Vet. Rec. 2003, 152, 323–329. [Google Scholar] [CrossRef]
  307. Aragon, C.L.; Hofmeister, E.H.; Budsberg, S.C. Systematic review of clinical trials of treatments for osteoarthritis in dogs. J. Am. Vet. Med. Assoc. 2007, 230, 514–521. [Google Scholar] [CrossRef]
  308. Gupta, R.C.; Canerdy, T.D.; Lindley, J.; Konemann, M.; Minniear, J.; Carroll, B.A.; Hendrick, C.; Goad, J.T.; Rohde, K.; Doss, R.; et al. Comparative therapeutic efficacy and safety of type-II collagen (uc-II), glucosamine and chondroitin in arthritic dogs: Pain evaluation by ground force plate. J. Anim. Physiol. Anim. Nutr. 2012, 96, 770–777. [Google Scholar] [CrossRef]
  309. Scott, R.M.; Evans, R.; Conzemius, M.G. Efficacy of an oral nutraceutical for the treatment of canine osteo arthritis. Vet. Comp. Orthop. Traumatol. 2017, 30, 318–323. [Google Scholar] [CrossRef] [PubMed]
  310. Shi, P.; Xu, S.; Yang, Z.; Wang, L.; Wu, Y.; Li, Y.; Zhu, Z. Harnessing gut microbiota for longevity: Insights into mechanisms and genetic manipulation. iMetaOmics 2024, 1, e36. [Google Scholar] [CrossRef]
  311. Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut Microbiota and Extreme Longevity. Curr. Biol. 2016, 26, 1480–1485. [Google Scholar] [CrossRef] [PubMed]
  312. Badal, V.D.; Vaccariello, E.D.; Murray, E.R.; Yu, K.E.; Knight, R.; Jeste, D.V.; Nguyen, T.T. The Gut Microbiome, Aging, and Longevity: A Systematic Review. Nutrients 2020, 12, 3759. [Google Scholar] [CrossRef]
  313. Sacoor, C.; Marugg, J.D.; Lima, N.R.; Empadinhas, N.; Montezinho, L. Gut-Brain Axis Impact on Canine Anxiety Disorders: New Challenges for Behavioral Veterinary Medicine. Vet. Med. Int. 2024, 2024, 2856759. [Google Scholar] [CrossRef]
  314. Tripathi, A.; Debelius, J.; Brenner, D.A.; Karin, M.; Loomba, R.; Schnabl, B.; Knight, R. The gut–liver axis and the intersection with the microbiome. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 397–411. [Google Scholar] [CrossRef]
  315. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain–gut–kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
  316. Marsland, B.J.; Trompette, A.; Gollwitzer, E.S. The Gut-Lung Axis in Respiratory Disease. Ann. Am. Thorac. Soc. 2015, 12 (Suppl. S2), S150–S156. [Google Scholar] [CrossRef]
  317. Margolis, K.G.; Cryan, J.F.; Mayer, E.A. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021, 160, 1486–1501. [Google Scholar] [CrossRef] [PubMed]
  318. Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The Brain-Gut-Microbiome Axis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef] [PubMed]
  319. Guo, Y.; Chen, X.; Gong, P.; Li, G.; Yao, W.; Yang, W. The Gut–Organ-Axis Concept: Advances the Application of Gut-on-Chip Technology. Int. J. Mol. Sci. 2023, 24, 4089. [Google Scholar] [CrossRef] [PubMed]
  320. Scriven, M.; Dinan, T.G.; Cryan, J.F.; Wall, M. Neuropsychiatric Disorders: Influence of Gut Microbe to Brain Signalling. Diseases 2018, 6, 78. [Google Scholar] [CrossRef]
  321. Doenyas, C.; Clarke, G.; Cserjési, R. Gut–brain axis and neuropsychiatric health: Recent advances. Sci. Rep. 2025, 15, 3415. [Google Scholar] [CrossRef]
  322. Choi, T.-Y.; Choi, Y.P.; Koo, J.W. Mental Disorders Linked to Crosstalk between The Gut Microbiome and The Brain. Exp. Neurobiol. 2020, 29, 403–416. [Google Scholar] [CrossRef]
  323. Auteri, M.; Zizzo, M.G.; Serio, R. GABA and GABA receptors in the gastrointestinal tract: From motility to inflammation. Pharmacol. Res. 2015, 93, 11–21. [Google Scholar] [CrossRef]
  324. Liu, L.; Huh, J.R.; Shah, K. Microbiota and the gut-brain-axis: Implications for new therapeutic design in the CNS. eBioMedicine 2022, 77, 103908. [Google Scholar] [CrossRef]
  325. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  326. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  327. Qu, S.; Yu, Z.; Zhou, Y.; Wang, S.; Jia, M.; Chen, T.; Zhang, X. Gut microbiota modulates neurotransmitter and gut-brain signaling. Microbiol. Res. 2024, 287, 127858. [Google Scholar] [CrossRef]
  328. Parashar, A.; Udayabanu, M. Gut microbiota regulates key modulators of social behavior. Eur. Neuropsychopharmacol. 2016, 26, 78–91. [Google Scholar] [CrossRef] [PubMed]
  329. Berger, M.; Gray, J.A.; Roth, B.L. The Expanded Biology of Serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef] [PubMed]
  330. Missale, C.; Nash, S.R.; Robinson, S.W.; Jaber, M.; Caron, M.G. Dopamine Receptors: From Structure to Function. Physiol. Rev. 1998, 78, 189–225. [Google Scholar] [CrossRef] [PubMed]
  331. Mohebi, A.; Pettibone, J.R.; Hamid, A.A.; Wong, J.-M.T.; Vinson, L.T.; Patriarchi, T.; Tian, L.; Kennedy, R.T.; Berke, J.D. Dissociable dopamine dynamics for learning and motivation. Nature 2019, 570, 65–70. [Google Scholar] [CrossRef]
  332. Cryan, J.F.; O’Mahony, S.M. The microbiome-gut-brain axis: From bowel to behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef]
  333. Luna, R.A.; Foster, J.A. Gut brain axis: Diet microbiota interactions and implications for modulation of anxiety and depression. Curr. Opin. Biotechnol. 2015, 32, 35–41. [Google Scholar] [CrossRef]
  334. Ambrosini, Y.M.; Borcherding, D.; Kanthasamy, A.; Kim, H.J.; Willette, A.A.; Jergens, A.; Allenspach, K.; Mochel, J.P. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front. Aging Neurosci. 2019, 11, 130. [Google Scholar] [CrossRef]
  335. Li, S.; Zhuo, M.; Huang, X.; Huang, Y.; Zhou, J.; Xiong, D.; Li, J.; Liu, Y.; Pan, Z.; Li, H.; et al. Altered gut microbiota associated with symptom severity in schizophrenia. PeerJ 2020, 8, e9574. [Google Scholar] [CrossRef] [PubMed]
  336. Malan-Müller, S.; Valles-Colomer, M.; Palomo, T.; Leza, J.C. The gut-microbiota-brain axis in a Spanish population in the aftermath of the COVID-19 pandemic: Microbiota composition linked to anxiety, trauma, and depression profiles. Gut Microbes 2023, 15, 2162306. [Google Scholar] [CrossRef] [PubMed]
  337. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [PubMed]
  338. Kim, C.-S.; Shin, G.-E.; Cheong, Y.; Shin, J.H.; Shin, D.-M.; Chun, W.Y. Experiencing social exclusion changes gut microbiota composition. Transl. Psychiatry 2022, 12, 254. [Google Scholar] [CrossRef]
  339. Silvestrino, M.; Pirolo, M.; Bianco, A.; Castellana, S.; Del Sambro, L.; Tarallo, V.D.; Guardabassi, L.; Zatelli, A.; Gernone, F. Idiopathic epilepsy in dogs is associated with dysbiotic faecal microbiota. Anim. Microbiome 2025, 7, 31. [Google Scholar] [CrossRef]
  340. Gernone, F.; Uva, A.; Silvestrino, M.; Cavalera, M.A.; Zatelli, A. Role of Gut Microbiota through Gut-Brain Axis in Epileptogenesis: A Systematic Review of Human and Veterinary Medicine. Biology 2022, 11, 1290. [Google Scholar] [CrossRef]
  341. Suchodolski, J.S.; Dowd, S.E.; Wilke, V.; Steiner, J.M.; Jergens, A.E. 16S rRNA Gene Pyrosequencing Reveals Bacterial Dysbiosis in the Duodenum of Dogs with Idiopathic Inflammatory Bowel Disease. PLoS ONE 2012, 7, e39333. [Google Scholar] [CrossRef]
  342. Suchodolski, J.S.; Markel, M.E.; Garcia-Mazcorro, J.F.; Unterer, S.; Heilmann, R.M.; Dowd, S.E.; Kachroo, P.; Ivanov, I.; Minamoto, Y.; Dillman, E.M.; et al. The Fecal Microbiome in Dogs with Acute Diarrhea and Idiopathic Inflammatory Bowel Disease. PLoS ONE 2012, 7, e51907. [Google Scholar] [CrossRef]
  343. Isaiah, A.; Parambeth, J.C.; Steiner, J.M.; Lidbury, J.A.; Suchodolski, J.S. The fecal microbiome of dogs with exocrine pancreatic insufficiency. Anaerobe 2017, 45, 50–58. [Google Scholar] [CrossRef]
  344. Breczko, W.J.; Bubak, J.; Miszczak, M. The Importance of Intestinal Microbiota and Dysbiosis in the Context of the Development of Intestinal Lymphoma in Dogs and Cats. Cancers 2024, 16, 2255. [Google Scholar] [CrossRef]
  345. Aziz, T.; Hussain, N.; Hameed, Z.; Lin, L. Elucidating the role of diet in maintaining gut health to reduce the risk of obesity, cardiovascular and other age-related inflammatory diseases: Recent challenges and future recommendations. Gut Microbes 2024, 16, 2297864. [Google Scholar] [CrossRef]
  346. Lee, A.H.; Lin, C.-Y.; Do, S.; Oba, P.M.; Belchik, S.E.; Steelman, A.J.; Schauwecker, A.; Swanson, K.S. Dietary supplementation with fiber, “biotics,” and spray-dried plasma affects apparent total tract macronutrient digestibility and the fecal characteristics, fecal microbiota, and immune function of adult dogs. J. Anim. Sci. 2022, 100, skac048. [Google Scholar] [CrossRef]
  347. Lin, C.Y.; Alexander, C.; Steelman, A.J.; Warzecha, C.M.; de Godoy, M.R.C.; Swanson, K.S. Effects of a Saccharomyces cerevisiae fermentation product on fecal characteristics, nutrient digestibility, fecal fermentative end-products, fecal microbial populations, immune function, and diet palatability in adult dogs1. J. Anim. Sci. 2019, 97, 1586–1599. [Google Scholar] [CrossRef]
  348. Finet, S.; He, F.; Clark, L.V.; de Godoy, M.R.C. Functional properties of miscanthus fiber and prebiotic blends in extruded canine diets. J. Anim. Sci. 2022, 100, skac078. [Google Scholar] [CrossRef] [PubMed]
  349. Song, H.; Lee, J.; Yi, S.; Kim, W.-H.; Kim, Y.; Namgoong, B.; Choe, A.; Cho, G.; Shin, J.; Park, Y.; et al. Red Ginseng Dietary Fiber Shows Prebiotic Potential by Modulating Gut Microbiota in Dogs. Microbiol. Spectr. 2023, 11, e00949-23. [Google Scholar] [CrossRef] [PubMed]
  350. Oba, P.M.; De La Guardia Hidrogo, V.M.; Kelly, J.; Saunders-Blades, J.; Steelman, A.J.; Swanson, K.S. Effects of diets supplemented with bioactive peptides on nutrient digestibility, immune cell responsiveness, and fecal characteristics, microbiota, and metabolites of adult cats. J. Anim. Sci. 2024, 102, skae104. [Google Scholar] [CrossRef] [PubMed]
  351. de Oliveira Matheus, L.F.; Risolia, L.W.; Ernandes, M.C.; de Souza, J.M.; Oba, P.M.; Vendramini, T.H.A.; Pedrinelli, V.; Henríquez, L.B.F.; de Oliveira Massoco, C.; Pontieri, C.F.F.; et al. Effects of Saccharomyces cerevisiae cell wall addition on feed digestibility, fecal fermentation and microbiota and immunological parameters in adult cats. BMC Vet. Res. 2021, 17, 351. [Google Scholar] [CrossRef]
  352. Xia, J.; Cui, Y.; Guo, Y.; Liu, Y.; Deng, B.; Han, S. The Function of Probiotics and Prebiotics on Canine Intestinal Health and Their Evaluation Criteria. Microorganisms 2024, 12, 1248. [Google Scholar] [CrossRef]
  353. Kim, D.-H.; Jeong, D.; Kang, I.-B.; Lim, H.-W.; Cho, Y.; Seo, K.-H. Modulation of the intestinal microbiota of dogs by kefir as a functional dairy product. J. Dairy Sci. 2019, 102, 3903–3911. [Google Scholar] [CrossRef]
  354. Wang, W.; Xu, L.; Cao, Y.; Liu, G.; Zhang, Y.; Wang, X.; Mao, X. Effects of supplementation with krill oil on blood parameters, hair quality, and fecal microbiota in male beagle dogs. Front. Microbiol. 2025, 16, 1587149. [Google Scholar] [CrossRef]
  355. Molina, R.A.; Villar, M.D.; Miranda, M.H.; Maldonado, N.C.; Vignolo, G.M.; Nader-Macias, M.E.F. A multi-strain probiotic promoted recovery of puppies from gastroenteritis in a randomized, double-blind, placebo-controlled study. Can. Vet. J. 2023, 64, 666–673. [Google Scholar]
  356. Rossi, G.; Cerquetella, M.; Gavazza, A.; Galosi, L.; Berardi, S.; Mangiaterra, S.; Mari, S.; Suchodolski, J.S.; Lidbury, J.A.; Steiner, J.M.; et al. Rapid Resolution of Large Bowel Diarrhea after the Administration of a Combination of a High-Fiber Diet and a Probiotic Mixture in 30 Dogs. Vet. Sci. 2020, 7, 21. [Google Scholar] [CrossRef] [PubMed]
  357. Nixon, S.L.; Rose, L.; Muller, A.T. Efficacy of an orally administered anti-diarrheal probiotic paste (Pro-Kolin Advanced) in dogs with acute diarrhea: A randomized, placebo-controlled, double-blinded clinical study. J. Vet. Int. Med. 2019, 33, 1286–1294. [Google Scholar] [CrossRef] [PubMed]
  358. Gómez-Gallego, C.; Junnila, J.; Männikkö, S.; Hämeenoja, P.; Valtonen, E.; Salminen, S.; Beasley, S. A canine-specific probiotic product in treating acute or intermittent diarrhea in dogs: A double-blind placebo-controlled efficacy study. Vet. Microbiol. 2016, 197, 122–128. [Google Scholar] [CrossRef] [PubMed]
  359. Atuahene, D.; Mukarram, S.A.; Balouei, F.; Antwi, A. Gut Health Optimization in Canines and Felines: Exploring the Role of Probiotics and Nutraceuticals. Pets 2024, 1, 135–151. [Google Scholar] [CrossRef]
  360. Kiełbik, P.; Witkowska-Piłaszewicz, O. The Relationship between Canine Behavioral Disorders and Gut Microbiome and Future Therapeutic Perspectives. Animals 2024, 14, 2048. [Google Scholar] [CrossRef]
  361. Voith, V.L. The impact of companion animal problems on society and the role of veterinarians. Vet. Clin. N. Am. Small Anim. Pract. 2009, 39, 327–345. [Google Scholar] [CrossRef]
  362. Powell, L.; Watson, B.; Serpell, J. Understanding feline feelings: An investigation of cat owners’ perceptions of problematic cat behaviors. Appl. Anim. Behav. Sci. 2023, 266, 106025. [Google Scholar] [CrossRef]
  363. Kubinyi, E.; Bel Rhali, S.; Sándor, S.; Szabó, A.; Felföldi, T. Gut Microbiome Composition is Associated with Age and Memory Performance in Pet Dogs. Animals 2020, 10, 1488. [Google Scholar] [CrossRef]
  364. Baik, J.-H. Dopamine Signaling in reward-related behaviors. Front. Neural Circuits 2013, 7, 152. [Google Scholar] [CrossRef]
  365. Jie, F.; Yin, G.; Yang, W.; Yang, M.; Gao, S.; Lv, J.; Li, B. Stress in Regulation of GABA Amygdala System and Relevance to Neuropsychiatric Diseases. Front. Neurosci. 2018, 12, 562. [Google Scholar] [CrossRef]
  366. Rosado, B.; García-Belenguer, S.; León, M.; Chacón, G.; Villegas, A.; Palacio, J. Blood concentrations of serotonin, cortisol and dehydroepiandrosterone in aggressive dogs. Appl. Anim. Behav. Sci. 2010, 123, 124–130. [Google Scholar] [CrossRef]
  367. León, M.; Rosado, B.; García-Belenguer, S.; Chacón, G.; Villegas, A.; Palacio, J. Assessment of serotonin in serum, plasma, and platelets of aggressive dogs. J. Vet. Behav. 2012, 7, 348–352. [Google Scholar] [CrossRef]
  368. González-Martínez, Á.; Muñiz de Miguel, S.; Graña, N.; Costas, X.; Diéguez, F.J. Serotonin and Dopamine Blood Levels in ADHD-Like Dogs. Animals 2023, 13, 1037. [Google Scholar] [CrossRef] [PubMed]
  369. Riva, J.; Bondiolotti, G.; Michelazzi, M.; Verga, M.; Carenzi, C. Anxiety related behavioural disorders and neurotransmitters in dogs. Appl. Anim. Behav. Sci. 2008, 114, 168–181. [Google Scholar] [CrossRef]
  370. Homer, B.; Judd, J.; Mohammadi Dehcheshmeh, M.; Ebrahimie, E.; Trott, D.J. Gut Microbiota and Behavioural Issues in Production, Performance, and Companion Animals: A Systematic Review. Animals 2023, 13, 1458. [Google Scholar] [CrossRef]
  371. Gorzelanna, Z.; Miszczak, M. Through the Intestines to the Head? That Is, How the Gastrointestinal Microbiota Affects the Behavior of Companion Animals. Pets 2024, 1, 201–215. [Google Scholar] [CrossRef]
  372. Cannas, S.; Tonini, B.; Belà, B.; Di Prinzio, R.; Pignataro, G.; Di Simone, D.; Gramenzi, A. Effect of a novel nutraceutical supplement (Relaxigen Pet dog) on the fecal microbiome and stress-related behaviors in dogs: A pilot study. J. Vet. Behav. 2021, 42, 37–47. [Google Scholar] [CrossRef]
  373. Roy, A.-S.; Aberkane, F.Z.; Cisse, S.; Guibert, A.; Richard, D.; Lerouzic, M.; Suor-cherer, S.; Boisard, S.; Guilet, D.; Benarbia, M.E.A.B.; et al. Metabolomics provides novel understanding of Melissa officinalis mechanism of action ensuring its calming effect on dogs. BMC Vet. Res. 2025, 21, 459. [Google Scholar] [CrossRef]
  374. Ozawa, M.; Inoue, M.; Uchida, K.; Chambers, J.K.; Takeuch, Y.; Nakayama, H. Physical signs of canine cognitive dysfunction. J. Vet. Med. Sci. 2019, 81, 1829–1834. [Google Scholar] [CrossRef]
  375. Landsberg, G.M.; Denenberg, S.; Araujo, J.A. Cognitive dysfunction in cats: A syndrome we used to dismiss as ‘old age’. J. Feline Med. Surg. 2010, 12, 837–848. [Google Scholar] [CrossRef]
  376. Sechi, S.; Chiavolelli, F.; Spissu, N.; Di Cerbo, A.; Canello, S.; Guidetti, G.; Fiore, F.; Cocco, R. An Antioxidant Dietary Supplement Improves Brain-Derived Neurotrophic Factor Levels in Serum of Aged Dogs: Preliminary Results. J. Vet. Med. 2015, 2015, 412501. [Google Scholar] [CrossRef]
  377. Reichling, J.; Frater-Schröder, M.; Herzog, K.; Bucher, S.; Saller, R. Reduction of behavioural disturbances in elderly dogs supplemented with a standardised Ginkgo leaf extract. Schweiz. Arch. Tierheilk. 2006, 158, 257–263. [Google Scholar] [CrossRef] [PubMed]
  378. Araujo, J.A.; Landsberg, G.M.; Milgram, N.W.; Miolo, A. Improvement of short-term memory performance in aged beagles by a nutraceutical supplement containing phosphatidylserine, Ginkgo biloba, vitamin E, and pyridoxine. Can. Vet. J. 2008, 49, 379–385. [Google Scholar] [PubMed]
  379. Pero, M.E.; Cortese, L.; Mastellone, V.; Tudisco, R.; Musco, N.; Scandurra, A.; D’Aniello, B.; Vassalotti, G.; Bartolini, F.; Lombardi, P. Effects of a Nutritional Supplement on Cognitive Function in Aged Dogs and on Synaptic Function of Primary Cultured Neurons. Animals 2019, 9, 393. [Google Scholar] [CrossRef] [PubMed]
  380. McGrath, A.P.; Horschler, D.J.; Hancock, L. Feline Cognition and the Role of Nutrition: An Evolutionary Perspective and Historical Review. Animals 2024, 14, 1967. [Google Scholar] [CrossRef]
  381. Hoffman, J.M.; Tolbert, M.K.; Promislow, D.E.L.; The Dog Aging Project Consortium. Demographic factors associated with joint supplement use in dogs from the Dog Aging Project. Front. Vet. Sci. 2022, 9, 906521. [Google Scholar] [CrossRef]
  382. Vandeweerd, J.M.; Coisnon, C.; Clegg, P.; Cambier, C.; Pierson, A.; Hontoir, F.; Saegerman, C.; Gustin, P.; Buczinski, S. Systematic review of efficacy of nutraceuticals to alleviate clinical signs of osteoarthritis. J. Vet. Int. Med. 2012, 26, 448–456. [Google Scholar] [CrossRef]
  383. Barbeau-Grégoire, M.; Otis, C.; Cournoyer, A.; Moreau, M.; Lussier, B.; Troncy, E. A 2022 Systematic Review and Meta-Analysis of Enriched Therapeutic Diets and Nutraceuticals in Canine and Feline Osteoarthritis. Int. J. Mol. Sci. 2022, 23, 10384. [Google Scholar] [CrossRef]
  384. Johnson, K.A.; Lee, A.H.; Swanson, K.S. Nutrition and nutraceuticals in the changing management of osteoarthritis for dogs and cats. J. Am. Vet. Med. Assoc. 2020, 256, 1335–1341. [Google Scholar] [CrossRef]
  385. Gildea, E.; Scales-Theobald, E.; Thompson, J.; Cook, A.; Forde, K.; Skingley, G.; Lawrie, S.; Williamson, N.; Panter, C. Development and validation of a quality of life and treatment satisfaction measure in canine osteoarthritis. Front. Vet. Sci. 2024, 11, 1377019. [Google Scholar] [CrossRef]
  386. Martinez, S.E.; Chen, Y.; Ho, E.A.; Martinez, S.A.; Davies, N.M. Pharmacological effects of a C-phycocyanin-based multicomponent nutraceutical in an in-vitro canine chondrocyte model of osteoarthritis. Can. J. Vet. Res. 2015, 79, 241–249. [Google Scholar]
  387. Beale, B.S. Use of nutraceuticals and chondroprotectants in osteoarthritic dogs and cats. Vet. Clin. N. Am. Small Anim. Pract. 2004, 34, 271–289, viii. [Google Scholar] [CrossRef]
  388. Mehler, S.J.; May, L.R.; King, C.; Harris, W.S.; Shah, Z. A prospective, randomized, double blind, placebo-controlled evaluation of the effects of eicosapentaenoic acid and docosahexaenoic acid on the clinical signs and erythrocyte membrane polyunsaturated fatty acid concentrations in dogs with osteoarthritis. Prostaglandins Leukot. Essent. Fat. Acids 2016, 109, 1–7. [Google Scholar] [CrossRef]
  389. Servet, E.; Biourge, V.; Marniquet, P. Dietary intervention can improve clinical signs in osteoarthritic dogs. J. Nutr. 2006, 136, 1995S–1997S. [Google Scholar] [CrossRef] [PubMed]
  390. Musco, N.; Vassalotti, G.; Mastellone, V.; Cortese, L.; Della Rocca, G.; Molinari, M.L.; Calabro, S.; Tudisco, R.; Cutrignelli, M.I.; Lombardi, P. Effects of a nutritional supplement in dogs affected by osteoarthritis. Vet. Med. Sci. 2019, 5, 325–335. [Google Scholar] [CrossRef] [PubMed]
  391. Roush, J.K.; Cross, A.R.; Renberg, W.C.; Dodd, C.E.; Sixby, K.A.; Fritsch, D.A.; Allen, T.A.; Jewell, D.E.; Richardson, D.C.; Leventhal, P.S.; et al. Evaluation of the effects of dietary supplementation with fish oil omega-3 fatty acids on weight bearing in dogs with osteoarthritis. J. Am. Vet. Med. Assoc. 2010, 236, 67–73. [Google Scholar] [CrossRef] [PubMed]
  392. Bauer, J.E. Therapeutic use of fish oils in companion animals. J. Am. Vet. Med. Assoc. 2011, 239, 1441–1451. [Google Scholar] [CrossRef]
  393. Comblain, F.; Serisier, S.; Barthelemy, N.; Balligand, M.; Henrotin, Y. Review of dietary supplements for the management of osteoarthritis in dogs in studies from 2004 to 2014. J. Vet. Pharmacol. Ther. 2016, 39, 1–15. [Google Scholar] [CrossRef]
  394. Fritsch, D.; Allen, T.A.; Dodd, C.E.; Jewell, D.E.; Sixby, K.A.; Leventhal, P.S.; Hahn, K.A. Dose-titration effects of fish oil in osteoarthritic dogs. J. Vet. Int. Med. 2010, 24, 1020–1026. [Google Scholar] [CrossRef]
  395. Corbee, R.J.; Barnier, M.M.; van de Lest, C.H.; Hazewinkel, H.A. The effect of dietary long-chain omega-3 fatty acid supplementation on owner’s perception of behaviour and locomotion in cats with naturally occurring osteoarthritis. J. Anim. Physiol. Anim. Nutr. 2013, 97, 846–853. [Google Scholar] [CrossRef]
  396. Eckert, T.; Jährling-Butkus, M.; Louton, H.; Burg-Roderfeld, M.; Zhang, R.; Zhang, N.; Hesse, K.; Petridis, A.K.; Kožár, T.; Steinmeyer, J.; et al. Efficacy of Chondroprotective Food Supplements Based on Collagen Hydrolysate and Compounds Isolated from Marine Organisms. Mar. Drugs 2021, 19, 542. [Google Scholar] [CrossRef] [PubMed]
  397. Stabile, M.; Girelli, C.R.; Lacitignola, L.; Samarelli, R.; Crovace, A.; Fanizzi, F.P.; Staffieri, F. 1H-NMR metabolomic profile of healthy and osteoarthritic canine synovial fluid before and after UC-II supplementation. Sci. Rep. 2022, 12, 19716. [Google Scholar] [CrossRef] [PubMed]
  398. Dobenecker, B.; Böswald, L.F.; Reese, S.; Steigmeier-Raith, S.; Trillig, L.; Oesser, S.; Schunck, M.; Meyer-Lindenberg, A.; Hugenberg, J. The oral intake of specific Bioactive Collagen Peptides (BCP) improves gait and quality of life in canine osteoarthritis patients—A translational large animal model for a nutritional therapy option. PLoS ONE 2024, 19, e0308378. [Google Scholar] [CrossRef]
  399. Manfredi, S.; Di Ianni, F.; Di Girolamo, N.; Canello, S.; Gnudi, G.; Guidetti, G.; Miduri, F.; Fabbi, M.; Daga, E.; Parmigiani, E.; et al. Effect of a commercially available fish-based dog food enriched with nutraceuticals on hip and elbow dysplasia in growing Labrador retrievers. Can. J. Vet. Res. 2018, 82, 154–158. [Google Scholar] [PubMed]
  400. Comblain, F.; Barthélémy, N.; Lefèbvre, M.; Schwartz, C.; Lesponne, I.; Serisier, S.; Feugier, A.; Balligand, M.; Henrotin, Y. A randomized, double-blind, prospective, placebo-controlled study of the efficacy of a diet supplemented with curcuminoids extract, hydrolyzed collagen and green tea extract in owner’s dogs with osteoarthritis. BMC Vet. Res. 2017, 13, 395. [Google Scholar] [CrossRef]
  401. Soontornvipart, K.; Wongsirichatchai, P.; Phongphuwanan, A.; Chatdarong, K.; Vimolmangkang, S. Cannabidiol plus krill oil supplementation improves chronic stifle osteoarthritis in dogs: A double-blind randomized controlled trial. Vet. J. 2024, 308, 106227. [Google Scholar] [CrossRef]
  402. Leong, D.J.; Gu, X.I.; Li, Y.; Lee, J.Y.; Laudier, D.M.; Majeska, R.J.; Schaffler, M.B.; Cardoso, L.; Sun, H.B. Matrix metalloproteinase-3 in articular cartilage is upregulated by joint immobilization and suppressed by passive joint motion. Matrix Biol. 2010, 29, 420–426. [Google Scholar] [CrossRef]
  403. Isaka, S.; Someya, A.; Nakamura, S.; Naito, K.; Nozawa, M.; Inoue, N.; Sugihara, F.; Nagaoka, I.; Kaneko, K. Evaluation of the effect of oral administration of collagen peptides on an experimental rat osteoarthritis model. Exp. Ther. Med. 2017, 13, 2699–2706. [Google Scholar] [CrossRef]
  404. Nakatani, S.; Mano, H.; Sampei, C.; Shimizu, J.; Wada, M. Chondroprotective effect of the bioactive peptide prolyl-hydroxyproline in mouse articular cartilage in vitro and in vivo. Osteoarthr. Cartil. 2009, 17, 1620–1627. [Google Scholar] [CrossRef]
  405. Stabile, M.; Fracassi, L.; Lacitignola, L.; Garcia-Pedraza, E.; Girelli, C.R.; Calculli, C.; D’Uggento, A.M.; Ribecco, N.; Crovace, A.; Fanizzi, F.P.; et al. Effects of a feed supplement, containing undenatured type II collagen (UC II®) and Boswellia Serrata, in the management of mild/moderate mobility disorders in dogs: A randomized, double-blind, placebo controlled, cross-over study. PLoS ONE 2024, 19, e0305697. [Google Scholar] [CrossRef]
  406. Martello, E.; Bigliati, M.; Adami, R.; Biasibetti, E.; Bisanzio, D.; Meineri, G.; Bruni, N. Efficacy of a dietary supplement in dogs with osteoarthritis: A randomized placebo-controlled, double-blind clinical trial. PLoS ONE 2022, 17, e0263971. [Google Scholar] [CrossRef]
  407. Malik, M.; Dixit, C.P.; Jacob, J.; Goswami, S. Chapter 17—Diseases of integument system of dogs and cats. In Introduction to Diseases, Diagnosis, and Management of Dogs and Cats; Rana, T., Ed.; Academic Press: Cambridge, MA, USA, 2024; pp. 257–270. [Google Scholar] [CrossRef]
  408. Saseendran, A.; Sherin K, G.; Banakar, P.; Rajkumar, G.; Ganapathy, J.; Sheethal, C. Skin Disease in Companion Animals: A Nutritional Impact. Indian J. Nat. Sci. (IJONS) 2016, 6, 10923–10931. [Google Scholar]
  409. Sun, X.; Ma, Y.; Gao, Y.; Li, J.; Li, Y.; Lv, L. Nutrients regulation of skin cells from canines and cats via Wnt/β-catenin signaling pathway. Front. Vet. Sci. 2025, 12, 1486201. [Google Scholar] [CrossRef] [PubMed]
  410. Watson, T.D. Diet and skin disease in dogs and cats. J. Nutr. 1998, 128, 2783S–2789S. [Google Scholar] [CrossRef] [PubMed]
  411. Combarros, D.; Castilla-Castaño, E.; Lecru, L.A.; Pressanti, C.; Amalric, N.; Cadiergues, M.C. A prospective, randomized, double blind, placebo-controlled evaluation of the effects of an n-3 essential fatty acids supplement (Agepi® ω3) on clinical signs, and fatty acid concentrations in the erythrocyte membrane, hair shafts and skin surface of dogs with poor quality coats. Prostaglandins Leukot. Essent. Fat. Acids 2020, 159, 102140. [Google Scholar] [CrossRef]
  412. Noli, C.; della Valle, M.F.; Miolo, A.; Medori, C.; Schievano, C.; The Skinalia Clinical Research Group. Effect of dietary supplementation with ultramicronized palmitoylethanolamide in maintaining remission in cats with nonflea hypersensitivity dermatitis: A double-blind, multicentre, randomized, placebo-controlled study. Vet. Dermatol. 2019, 30, 387-e117. [Google Scholar] [CrossRef]
  413. Kim, H.; Rather, I.A.; Kim, H.; Kim, S.; Kim, T.; Jang, J.; Seo, J.; Lim, J.; Park, Y.-H. A Double-Blind, Placebo Controlled-Trial of a Probiotic Strain Lactobacillus sakei Probio-65 for the Prevention of Canine Atopic Dermatitis. J. Microbiol. Biotechnol. 2015, 25, 1966–1969. [Google Scholar] [CrossRef]
  414. Song, H.; Mun, S.-H.; Han, D.-W.; Kang, J.-H.; An, J.-U.; Hwang, C.-Y.; Cho, S. Probiotics ameliorate atopic dermatitis by modulating the dysbiosis of the gut microbiota in dogs. BMC Microbiol. 2025, 25, 228. [Google Scholar] [CrossRef]
  415. Amundson, L.A.; Millican, A.A.; Swensson, E.; McGilliard, M.L.; Tomlinson, D. Effect of Supplemental Trace Mineral Source on Haircoat and Activity Levels in Senior Dogs. Animals 2025, 15, 686. [Google Scholar] [CrossRef]
  416. Kirby, N.A.; Hester, S.L.; Rees, C.A.; Kennis, R.A.; Zoran, D.L.; Bauer, J.E. Skin surface lipids and skin and hair coat condition in dogs fed increased total fat diets containing polyunsaturated fatty acids. J. Anim. Physiol. Anim. Nutr. 2009, 93, 505–511. [Google Scholar] [CrossRef] [PubMed]
  417. Wang, W.; Dong, H.; Chang, X.; Chen, Q.; Wang, L.; Chen, S.; Chen, L.; Wang, R.; Ge, S.; Wang, P.; et al. Bifidobacterium lactis and Lactobacillus plantarum Enhance Immune Function and Antioxidant Capacity in Cats through Modulation of the Gut Microbiota. Antioxidants 2024, 13, 764. [Google Scholar] [CrossRef] [PubMed]
  418. Olivry, T.; Mueller, R.S. Critically appraised topic on adverse food reactions of companion animals (3): Prevalence of cutaneous adverse food reactions in dogs and cats. BMC Vet. Res. 2017, 13, 51. [Google Scholar] [CrossRef] [PubMed]
  419. Vogelnest, L.; Cheng, K. Cutaneous adverse food reactions in cats: Retrospective evaluation of 17 cases in a dermatology referral population (2001–2011). Aust. Vet. J. 2013, 91, 443–451. [Google Scholar] [CrossRef]
  420. Gaschen, F.P.; Merchant, S.R. Adverse Food Reactions in Dogs and Cats. Vet. Clin. Small Anim. Pract. 2011, 41, 361–379. [Google Scholar] [CrossRef]
  421. Scrimshaw, N.S.; Suskind, R.M. Interactions of Nutrition and Infection. Dent. Clin. N. Am. 1976, 20, 461–472. [Google Scholar] [CrossRef]
  422. Munteanu, C.; Schwartz, B. The relationship between nutrition and the immune system. Front. Nutr. 2022, 9, 1082500. [Google Scholar] [CrossRef]
  423. Wu, D.; Lewis, E.D.; Pae, M.; Meydani, S.N. Nutritional Modulation of Immune Function: Analysis of Evidence, Mechanisms, and Clinical Relevance. Front. Immunol. 2019, 9, 3160. [Google Scholar] [CrossRef]
  424. Calder, P.C.; Carr, A.C.; Gombart, A.F.; Eggersdorfer, M. Optimal Nutritional Status for a Well-Functioning Immune System Is an Important Factor to Protect against Viral Infections. Nutrients 2020, 12, 1181. [Google Scholar] [CrossRef]
  425. Medoro, A.; Davinelli, S.; Colletti, A.; Di Micoli, V.; Grandi, E.; Fogacci, F.; Scapagnini, G.; Cicero, A.F.G. Nutraceuticals as Modulators of Immune Function: A Review of Potential Therapeutic Effects. Prev. Nutr. Food Sci. 2023, 28, 89–107. [Google Scholar] [CrossRef]
  426. Wang, W.; Xu, L.; Zhang, Y.; Cao, Y.; Yang, Y.; Liu, G.; Mao, X. Effects of Chenpi (Citrus reticulata cv. Chachiensis) on serum antioxidant enzymes, inflammatory factors, and intestinal health in Beagle dogs. Front. Microbiol. 2025, 15, 1415860. [Google Scholar] [CrossRef]
  427. Stuyven, E.; Verdonck, F.; Van Hoek, I.; Daminet, S.; Duchateau, L.; Remon, J.P.; Goddeeris, B.M.; Cox, E. Oral administration of beta-1,3/1,6-glucan to dogs temporally changes total and antigen-specific IgA and IgM. Clin. Vaccine Immunol. 2010, 17, 281–285. [Google Scholar] [CrossRef] [PubMed]
  428. Ferreira, L.G.; Endrighi, M.; Lisenko, K.G.; de Oliveira, M.R.D.; Damasceno, M.R.; Claudino, J.A.; Gutierres, P.G.; Peconick, A.P.; Saad, F.M.d.O.B.; Zangeronimo, M.G. Oat beta-glucan as a dietary supplement for dogs. PLoS ONE 2018, 13, e0201133. [Google Scholar] [CrossRef]
  429. Rentas, M.F.; Pedreira, R.S.; Perini, M.P.; Risolia, L.W.; Zafalon, R.V.A.; Alvarenga, I.C.; Vendramini, T.H.A.; Balieiro, J.C.C.; Pontieri, C.F.F.; Brunetto, M.A. Galactoligosaccharide and a prebiotic blend improve colonic health and immunity of adult dogs. PLoS ONE 2020, 15, e0238006. [Google Scholar] [CrossRef] [PubMed]
  430. Luo, J.B.; Zhang, L.; Fu, M.; Hong, Y.; Du, X.Y.; Cheng, G.Q.; Xia, J.Y.; Dong, H. Astragalus polysaccharide (APS) supplement in beagle dogs after castration: Effects on the haematology and serum chemistry profiles, immune response, and oxidative stress status. Vet. Med. Sci. 2023, 9, 98–110. [Google Scholar] [CrossRef] [PubMed]
  431. Kayser, E.; Castaneda, P.L.; Soto-Diaz, K.; Steelman, A.J.; Murphy, A.; Spindola, M.; He, F.; de Godoy, M.R.C. Functional properties of Ganoderma lucidum supplementation in canine nutrition. J. Anim. Sci. 2024, 102, skae051. [Google Scholar] [CrossRef]
  432. Rutherfurd-Markwick, K.J.; Hendriks, W.H.; Morel, P.C.H.; Thomas, D.G. The potential for enhancement of immunity in cats by dietary supplementation. Vet. Immunol. Immunopathol. 2013, 152, 333–340. [Google Scholar] [CrossRef]
  433. Rossi, G.; Pengo, G.; Galosi, L.; Berardi, S.; Tambella, A.M.; Attili, A.R.; Gavazza, A.; Cerquetella, M.; Jergens, A.E.; Guard, B.C.; et al. Effects of the Probiotic Mixture Slab51® (SivoMixx®) as Food Supplement in Healthy Dogs: Evaluation of Fecal Microbiota, Clinical Parameters and Immune Function. Front. Vet. Sci. 2020, 7, 613. [Google Scholar] [CrossRef]
  434. Guidetti, G.; Di Cerbo, A.; Giovazzino, A.; Rubino, V.; Palatucci, A.T.; Centenaro, S.; Fraccaroli, E.; Cortese, L.; Bonomo, M.G.; Ruggiero, G.; et al. In Vitro Effects of Some Botanicals with Anti-Inflammatory and Antitoxic Activity. J. Immunol. Res. 2016, 2016, 5457010. [Google Scholar] [CrossRef]
  435. Di Cerbo, A.; Scarano, A.; Pezzuto, F.; Guidetti, G.; Canello, S.; Pinetti, D.; Genovese, F.; Corsi, L. Oxytetracycline-Protein Complex: The Dark Side of Pet Food. Open Public Health J. 2018, 11, 162–169. [Google Scholar] [CrossRef]
  436. Di Cerbo, A.; Canello, S.; Guidetti, G.; Fiore, F.; Corsi, L.; Rubattu, N.; Testa, C.; Cocco, R. Adverse food reactions in dogs due to antibiotic residues in pet food: A preliminary study. Vet. Ital. 2018, 54, 137–146. [Google Scholar] [CrossRef]
  437. Di Cerbo, A.; Pezzuto, F.; Guidetti, G.; Canello, S.; Corsi, L. Tetracyclines: Insights and Updates of their Use in Human and Animal Pathology and their Potential Toxicity. Open Biochem. J. 2019, 13, 1–12. [Google Scholar] [CrossRef]
  438. Segarra, S.; Miró, G.; Montoya, A.; Pardo-Marín, L.; Teichenné, J.; Ferrer, L.; Cerón, J.J. Prevention of disease progression in Leishmania infantum-infected dogs with dietary nucleotides and active hexose correlated compound. Parasites Vectors 2018, 11, 103. [Google Scholar] [CrossRef]
  439. Meydani, S.N.; Wu, D.; Santos, M.S.; Hayek, M.G. Antioxidants and immune response in aged persons: Overview of present evidence. Am. J. Clin. Nutr. 1995, 62, 1462S–1476S. [Google Scholar] [CrossRef]
  440. Perondi, F.; Bisanzio, D.; Adami, R.; Lippi, I.; Meineri, G.; Cutrignelli, M.I.; Massa, S.; Martello, E. The effect of a diet supplement containing S-acetyl-glutathione (SAG) and other antioxidant natural ingredients on glutathione peroxidase in healthy dogs: A pilot study. Ital. J. Anim. Sci. 2023, 22, 589–593. [Google Scholar] [CrossRef]
Table 1. Nutraceutical substances effects in dogs and cats.
Table 1. Nutraceutical substances effects in dogs and cats.
Nutraceutical SubstanceMechanism of ActionClinical OutcomeAnimal ModelContribution to
Longevity
Omega-3 Fatty Acids
  • Anti-inflammatory and immune-modulating properties (IL-6, TNF-α, and eicosanoids reduction; impairment of leukocyte adhesion to endothelium, production of specific metabolites, competition with AA) [133,134,135,136,137,138,139,142,143,144]
  • Cardiovascular function improvement (modulation of ionic currents in cardiac cells, reduction in leukocytic infiltration in ischemic tissue) [146,147,148]
  • Positive influence on reproduction (modulation of molecules involved in reproduction, influence on sperm, oocytes, and corpus luteum development and functionality, improvement of semen quality) [149,150,151]
  • Renal protection (modulation of cholesterolemia, triglyceridemia, GFR, oxidative damage, inflammation) [152,153]
  • Neuroprotective and synaptogenesis promotion (support of brain development and, neurogenesis and glucose transport enhancement) [154,155,156]
  • Mobility improvement, pain relief and NSAID need reduction [140]
  • Skin and coat quality improvement [354]
  • Cardiovascular and renal health promotion [146,147,148]
  • Reproductive functions and semen quality enhancement [49,151]
  • Cognitive and vision support [154,155,156]
  • Joint conditions improvement and OA-related symptoms reduction [246,391,392,393,394]
  • Drooling, back and neck intense itching, neck eczema, chronic conjunctivitis and stomatitis improvement [42]
  • Dogs and cats with OA [246,391,392,393,394]
  • Dogs with OA [140]
  • Healthy dogs [354]
  • Cats with CAFR [42]
  • Dogs with ischemia-induced fatal cardiac ventricular arrhythmias [146]
  • dogs suffering from infertility associated with hypospermia [49]
  • Diseases prevention and chronic diseases progression slowing
  • Quality of life improvement
Prebiotics and Probiotics
  • Gut microbiota balance and SCFA production [161,176,177]
  • Disorders prevention or management (obesity, diabetes mellitus, gastrointestinal disorders) [178,179,180,181,190,191,192,193,194]
  • Pathogen inhibition (bacteriocins, competition for nutrients) [182]
  • Intestinal barrier strengthening (modulation of mucin and pro-inflammatory cytokines production) [183,184]
  • Stool features improvement [185,186,187,188,189]
  • Behavior modulation (influence on GBA) [195,199,200]
  • Gut–brain axis modulation (serotonin, GABA, dopamine, glutamate, signaling molecules) [324,325,326,327]
  • Stool quality improvement, beneficial shifting to fecal microbiota and metabolite profiles, blood lipids, and increased fecal IgA reduction [346]
  • Gastrointestinal and immune health, as well as fecal quality metabolites improvement [343]
  • Immune functions and antioxidant defense improvement [417,429,430,431,432,433,434]
  • Overall physical condition, hair, and fecal quality improvement in cats [417]
  • CKD markers improvement (BUN and creatinine concentrations reduction or preservation) and clinical symptoms improvement (appetite, activity, defecation frequency) [185]
  • Stomatitis, gingivostomatitis, and oral infections reduction [193]
  • Aggressiveness and anxiety decrease [191,199,200]
  • Gastrointestinal pathologies (gastroenteritis and diarrhea) fast recovery [183,351,352,353,354]
  • CADESI and PVAS score improvement [413,414]
  • Leishmaniosis progression slowing [430]
  • Gut microbiota improvement in dogs [344,349,350]
  • Cats with CKD at stage 2-3 [185]
  • Dogs suffering from anxiety and aggression [191,199,200,372]
  • Dogs suffering from exocrine pancreatic insufficiency [343]
  • healthy household dogs [349], and cats [350]
  • Dog puppies with gastroenteritis [351] and adults with diarrhea [183,352,353,354]
  • Healthy adult dogs [346,347,429,431]
  • Healthy cats [417,432]
  • Dogs suffering from atopic dermatitis [413,414]
  • Healthy adult castrated dogs [430]
  • Leishmania infantum-infected dogs [434]
  • Gastrointestinal and extraintestinal health support and improvement
  • Immunity status strengthening
  • Chronic diseases progression slowing
Plant Extracts
  • Antioxidant and anti-inflammatory activity (TNF-α, IL-1β, and COX-2 reduction) [97,210,211,212,213,214,218]
  • Immunomodulation [76,211]
  • Antimicrobial and detoxifying activity [210,211]
  • Cardioprotection and anticancer [76,210,211]
  • Lameness, willingness to move, play, and jump improvement [405,406]
  • Intermittent lameness, local pain and stiff gait reduction in severity and resolution [225],
  • Stress reduction [53]
  • Metabolic profile improvement [104]
  • lesions associated with granulomatous dermatitis (papules, nodules, plaques) and external otitis reduction [43]
  • Behavioral issues (marking, anxiety, diffidence, irregular biorhythm, reactivity, activation, irritability, alertness, environmental exploration and attention requirement, fear, and hyperactivity) reduction [47,48]
  • Antioxidant and immunological functions [354,435] enhancement
  • CKS [44], epiphora [45], and leishmaniosis [52] progression slowing
  • Healthy dogs [104]
  • Dogs with behavioral issues [47,48]
  • Dogs suffering from chronic joint and spinal disease [225]
  • Dogs suffering from mobility problems [405,406]
  • dogs suffering from granulomatous dermatitis and external otitis [43]
  • Adult and healthy dogs [354,435]
  • Dogs suffering from CKS [44] and epiphora [45], infected by Leishmania infantum [52]
  • Quality of life improvement
  • Immunity status strengthening
Dietary supplements
  • Anti-inflammatory and antioxidant defense (e.g., radical scavenging activity, synergism with other antioxidant molecules, influence of the activity of antioxidant enzymes, such as SOD) [224,228,231,234,290,292,301]
  • Tissues and organs development [224,228,229]
  • Coagulation process improvement [232,233]
  • Cofactors or coenzymes in the metabolism of carbohydrates, proteins and lipids [225,226,227]
  • Ensure the proper functioning of Antioxidant enzymes (SOD, alkaline phosphatase) [243,244]
  • Ensure skeletal health and proper development of bones [251,252,253]
  • Cancer prevention [262,263,264]
  • Intracellular antioxidant, redox balance, maintenance, cofactors of antioxidant and detoxification enzymes [271,273,274,275]
  • Toxic compounds and xenobiotics detoxification [269,272,286]
  • Hepatoprotection [287]
  • Cartilage protection [288,293]
  • Vision improvement [224]
  • Skeletal and immune health promotion [229,230,241,242]
  • Antioxidant defense improvement [433]
  • Hair growth increase, superior hair quality, hair shedding decrease, and more hours of activity per day increase [415]
  • Oxidative stress and related disease protection (hepatic, endocrine, GI) [259]
  • Significant increases in erythrocyte total and reduced glutathione [277]
  • SOD, catalase, glutathione peroxidase, total antioxidant capacity improvement [276]
  • Xenobiotics detoxification (e.g., conjugation with GSH, enhancement of the excretion of toxic compounds from cells/body, direct neutralization of toxic compounds) [269,272]
  • GSH erythrocytic levels and liver parameters (ALT, AST, ALP, GGT, BIL) improvement [287]
  • scores for pain, weight-bearing and osteoarthritis severity reduction [304]
  • Glycosaminoglycan synthesis increase and cartilage-degrading enzymes inhibition [288]
  • Acetaminophen-induced toxicity reduction in cats [286]
  • Cats intoxicated with acetaminophen [286]
  • Dogs suffering from liver disease [290]
  • Dogs suffering from OA [304]
  • Healthy dogs [438]
  • Healthy senior dogs [415]
  • Cats suffering from clinically evident and subclinical hypertrophic cardiomyopathy [276]
  • Healthy senior cats [277]
  • Quality of life improvement
  • Antioxidant defense improvement
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nicotra, M.; Iannitti, T.; Di Cerbo, A. Nutraceuticals, Social Interaction, and Psychophysiological Influence on Pet Health and Well-Being: Focus on Dogs and Cats. Vet. Sci. 2025, 12, 964. https://doi.org/10.3390/vetsci12100964

AMA Style

Nicotra M, Iannitti T, Di Cerbo A. Nutraceuticals, Social Interaction, and Psychophysiological Influence on Pet Health and Well-Being: Focus on Dogs and Cats. Veterinary Sciences. 2025; 12(10):964. https://doi.org/10.3390/vetsci12100964

Chicago/Turabian Style

Nicotra, Mario, Tommaso Iannitti, and Alessandro Di Cerbo. 2025. "Nutraceuticals, Social Interaction, and Psychophysiological Influence on Pet Health and Well-Being: Focus on Dogs and Cats" Veterinary Sciences 12, no. 10: 964. https://doi.org/10.3390/vetsci12100964

APA Style

Nicotra, M., Iannitti, T., & Di Cerbo, A. (2025). Nutraceuticals, Social Interaction, and Psychophysiological Influence on Pet Health and Well-Being: Focus on Dogs and Cats. Veterinary Sciences, 12(10), 964. https://doi.org/10.3390/vetsci12100964

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