A one-health approach to identifying and mitigating the impact of endocrine disorders on human and equine athletes

Jane M. Manfredi Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI

Search for other papers by Jane M. Manfredi in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
,
Sarah I. Jacob Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI

Search for other papers by Sarah I. Jacob in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
,
Brooke L. Boger Comparative Medicine and Integrative Biology, Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI

Search for other papers by Brooke L. Boger in
Current site
Google Scholar
PubMed
Close
 BA
, and
Elaine M. Norton Department of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ

Search for other papers by Elaine M. Norton in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
Open access

Abstract

Endocrinopathies affect multiple species in ever-increasing percentages of their populations, creating an opportunity to apply one-health approaches to determining creative preventative measures and therapies in athletes. Obesity and alterations in insulin and glucose dynamics are medical concerns that play a role in whole-body health and homeostasis in both horses and humans. The role and impact of endocrine disorders on the musculoskeletal, cardiovascular, and reproductive systems are of particular interest to the athlete. Elucidation of both physiologic and pathophysiologic mechanisms involved in disease processes, starting in utero, is important for development of prevention and treatment strategies for the health and well-being of all species. This review focuses on the unrecognized effects of endocrine disorders associated with the origins of metabolic disease; inflammation at the intersection of endocrine disease and related diseases in the musculoskeletal, cardiovascular, and reproductive systems; novel interventions; and diagnostics that are informed via multiomic and one-health approaches. Readers interested in further details on specific equine performance conditions associated with endocrine disease are invited to read the companion Currents in One Health by Manfredi et al, JAVMA, February 2023.

Abstract

Endocrinopathies affect multiple species in ever-increasing percentages of their populations, creating an opportunity to apply one-health approaches to determining creative preventative measures and therapies in athletes. Obesity and alterations in insulin and glucose dynamics are medical concerns that play a role in whole-body health and homeostasis in both horses and humans. The role and impact of endocrine disorders on the musculoskeletal, cardiovascular, and reproductive systems are of particular interest to the athlete. Elucidation of both physiologic and pathophysiologic mechanisms involved in disease processes, starting in utero, is important for development of prevention and treatment strategies for the health and well-being of all species. This review focuses on the unrecognized effects of endocrine disorders associated with the origins of metabolic disease; inflammation at the intersection of endocrine disease and related diseases in the musculoskeletal, cardiovascular, and reproductive systems; novel interventions; and diagnostics that are informed via multiomic and one-health approaches. Readers interested in further details on specific equine performance conditions associated with endocrine disease are invited to read the companion Currents in One Health by Manfredi et al, JAVMA, February 2023.

Endocrinopathies, with their various systems interconnections, have the ability to greatly affect athletic performance in multiple species. Recognizing the similarities between human and equine endocrine disorders (Figure 1) allows us to take a one-health approach to investigate the potential impact of endocrinopathies on the musculoskeletal, cardiovascular, and reproductive systems in athletes. Human metabolic syndrome (MetS) is a consortium of disorders that affects 25% of the United States population and is a major risk factor for development of obesity, diabetes, cardiovascular disease, and orthopedic disease.13 Endocrine disease is also prevalent in the horse population, with 21% of horses over 15 years old diagnosed with pituitary pars intermedia dysfunction (PPID) and an estimated 20 to 40% of the equine population being at risk for the development of equine metabolic syndrome (EMS).4 Details of the impact of PPID, EMS, and the insulin dysregulation (ID) that can accompany them in body systems and conditions other than laminitis are limited. However, the unrecognized effects of endocrine disorders, which will be the focus of this article, include suspensory ligament degeneration, lameness, osteoarthritis (OA), developmental orthopedic disease, muscle atrophy, cardiovascular disease, placental dysfunction, and infertility.512 Future research collating phenotype to multiomic analyses (genomics, metabolomics, transcriptomics, proteomics, and microbiomics; Figure 2) and examination of the developmental origin of metabolic disease to evaluate the links between endocrinopathies and athletic performance can contribute to our understanding of the pathophysiology, while shedding light on novel biomarkers and treatments.

Figure 1
Figure 1

Shared systemic effects of endocrine disease in the horse and human. Created with BioRender.com.

Citation: American Journal of Veterinary Research 84, 2; 10.2460/ajvr.22.11.0194

Figure 2
Figure 2

A combined approach to assessing phenotype in light of multiomic analyses (genomics, metabolomics, transcriptomics, proteomics, and microbiomics) to understand the effects of endocrine disorders on performance. Created with BioRender.com.

Citation: American Journal of Veterinary Research 84, 2; 10.2460/ajvr.22.11.0194

Current Understanding of the Pathophysiology of Insulin Dysregulation

ID, defined as disruptions in the interconnected relationships between insulin, glucose, and lipid metabolism, plays an important role in metabolic syndrome and contributes to the pathology associated with other athletic performance-limiting conditions.13 In normal conditions, insulin is released by β-cells from pancreatic islets of Langerhans primarily in response to hyperglycemia, but other macronutrients, hormones, and neurotransmitters can also stimulate insulin release.14 Glucose readily crosses the β-cell membrane via high-capacity, low-affinity glucose transporters (GLUT).15 Although GLUT2 is the primary β-cell transporter in the mouse, GLUT1 and GLUT3 appear to be more important in humans and the horse β-cell transporter has yet to be identified.16 In rodent models and humans, insulin secretion is biphasic with an initial rapid release followed by a more prolonged but less concentrated release;17 however, it has not been determined as to whether insulin release is mono or biphasic in horses.18,19

In the periphery, insulin mediates its effects by binding to insulin receptors located on its main target tissues: adipocytes, skeletal muscle, and liver. This binding activates intracellular insulin-responsive substrates (IRS), which are responsible for insulin’s metabolic, vascular, and mitogenic effects. For example, IRS activation of the downstream effector pathway phosphatidylinositol 3-kinase (PI3K) promotes insulin’s metabolic effects including the translocation of intracellular glucose transporter proteins (GLUT4) to the cell membrane, stimulating lipid and protein synthesis, and inhibiting glycogenolysis and hepatic gluconeogenesis.20 Once the glucose transporter is translocated to the plasma membrane, glucose is transported into the cell where it is phosphorylated to be stored as glycogen or utilized for adenosine triphosphate (ATP) production.

Hyperinsulinemia and insulin resistance are the mainstays of MetS. With insulin resistance, as blood glucose levels fail to return to normoglycemia, the pancreatic β-cells hyperfunction by increasing insulin secretion via β-cell hypertrophy and decreasing β-cell expression of glucokinase while increasing expression of hexokinase, shifting the insulin-glucose response curve to the right.21 In addition, in normal conditions, the first portal passage through the liver is responsible for 50 to 60% of insulin clearance in humans22 and up to 70% in horses;23 however, insulin insensitivity markedly reduces hepatic insulin clearance across species,23,24 contributing to hyperinsulinemia.

Incretin response and oral therapies

Postprandial stimulation of insulin release via the enteroinsular axis has been found to result in a greater insulin response versus intravenous or intraperitoneal glucose administration. This has been deemed the incretin response based on the effects of the incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1). Breed differences in GLP-1 concentrations were identified in ponies and Andalusians, 2 breeds considered high risk for EMS, who had higher GLP-1 and insulin concentrations compared to Standardbred horses, potentially indicating a risk factor for ID.25 Ponies with ID had hyperinsulinemia and an exaggerated incretin response 4 hours after consumption of pasture compared to controls.26 In contrast, other studies27 found that both active GLP-1 and total GLP-1 levels were decreased in horses with ID compared to controls.

Drugs that specifically target incretins are becoming more prominent. In humans, synthetic GLP-1 and drugs targeted at inhibiting dipeptidyl peptidase 4, a serine protease that inactivates GIP and GLP-1, have been shown to increase glycemic control in humans with MetS while also decreasing body weight.28 In a small cohort of horses with mild ID, a single dose of exenatide, a synthetic GLP-1 receptor agonist (also known as an incretin mimetic), resulted in a significant decrease in area under the curve of plasma insulin concentrations and improved insulin sensitivity after an oral sugar test.29 This seemingly contradictory result was postulated to be due to slower gastric emptying, thus decreasing the rate of glucose absorption or other independent actions that achieved increased insulin sensitivity.

Traditional dietary medications and supplements for treating EMS (above and beyond exercise and dietary change) include metformin, levothyroxine, and a resveratrol + amino acid blend, with the first and last also being used for humans with MetS.3041 However, a new promising class of oral treatment options, sodium-glucose cotransporter 2 inhibitors (ertugliflozin and canagliflozin), which modulate the sodium and glucose transport in the kidney by increasing urinary glucose excretion, have been described. These drugs have been shown to have efficacy in treating refractive cases of EMS/ID, ones where the typical diet, exercise, and supplementation with metformin or levothyroxine has failed; limit insulin peaks after eating; and improve Obel scores.42,43 Further investigation is required to determine if these new potential therapeutics are successful in a larger number of clinical cases and compared to other treatments.

Dyslipidemia

Dyslipidemia is a central component of the pathophysiology behind metabolic syndrome.44 Direct inhibition of insulin action by elevated fatty acids eliminates the negative feedback on hormone-sensitive lipase, leading to further accumulation of triglycerides in the liver and ectopic depositions of fat in muscle (intramyocellular lipid), reducing the cellular uptake of glucose, promoting hyperglycemia, and contributing to insulin resistance.45,46

In horses, elevated nonesterified fatty acids (NEFAs) and triglycerides have been associated with EMS, although not consistently across studies. Increased triglyceride levels were found in ponies with EMS, and hypertriglyceridemia was identified as a risk factor for the development of laminitis.47,48 Another study49 identified a seasonal elevation in triglyceride levels in ponies with EMS. In a cohort of large-breed horses, a significant elevation in NEFA concentrations was noted; however, triglyceride concentrations were not different.50 These studies reflect possible differences between ponies and horses with EMS. In a small cohort of obese large-breed horses with insulin resistance compared with metabolically healthy horses, the plasma concentration of very low-density lipoproteins and high-density lipoproteins (HDL) were found to be increased in horses with insulin resistance.50 The elevation in HDL is opposite to what is characteristically seen in humans with MetS and may reflect the absence of plasma cholesteryl ester transfer protein in equids.50 Using continuous lipid profile measurements, subfractions of HDL, specifically HDL3a, were found to be significantly lower in healthy horses compared to horses with obesity, laminitis, or both despite there being no difference in total HDL concentrations, possibly indicating a novel method of lipoprotein profiling.51 Unfortunately, this study51 did not evaluate metabolic status and further studies need to be performed to determine the utility of EMS.

Inflammatory cytokines

Evidence supports that a large contribution to the pathophysiology of metabolic syndrome is related to the role of adipose tissue in promoting chronic, low-grade inflammation. Adipose tissue is a biologically active endocrine organ that secretes a myriad of substances including cytokines, eicosanoids, complement proteins, binding proteins, vasoactive factors, and regulators of lipid metabolism that are collectively known as adipokines.52 The exact mechanism behind adipose-induced inflammation in metabolic syndrome is unknown, but it is proposed that “sick fat” is a result of adipose hypertrophy and hyperplasia secondary to excess nutrition leading to endoplasmic reticulum stress or hypoxia from an insufficient blood supply,53,54 which results in macrophage and mast cell infiltration and the production of inflammatory mediators.54,55 In humans, this theory is supported as certain adipose tissue depots (the subcutaneous and visceral abdominal depots) more commonly associated with MetS contain hypertrophied adipocytes, which undergo lipoapoptosis releasing inflammatory mediators.56 Additional hypotheses have proposed that endothelial stress and hyperactive platelets are the primary etiology behind adipose tissue inflammation as well as the low-level inflammation observed in other tissues.57 Regardless, in humans, the increased production of inflammatory mediators is a risk factor for type II diabetes mellitus58 and is associated with higher fasting insulin levels and dyslipidemia.

The interrelationship of inflammatory mediators in obesity and ID has also been investigated in horses with EMS. Similarly to humans, histological examination of adipose tissue in obese, hyperinsulinemic ponies revealed a marked degree of hypertrophy56,59,60 and macrophage infiltration59 compared to the adipose tissue of non-obese ponies, indicating adipose dysfunction. Other early support for adipose tissue inflammation in driving disease processes is the clinical improvement of a severely insulin-resistant animal that improved greatly after systemic injection with autologous 5-azacytadine-treated and resveratrol-treated (an anti-inflammatory agent)61 adipose stem cells.62 Studies56,60,6365 evaluating inflammatory cytokine mRNA or protein expression levels have identified significant differences between nuchal ligament, abdominal, and subcutaneous fat, suggesting that adipose tissue depots have unique biological behavior as also seen in humans. Horses with EMS were found to have marked increases in TNFα, interleukin-1β (IL-1β), and CCL2 in both perirenal and retroperitoneal fat depots,60 and IL-6 was significantly increased in the subcutaneous fat.59 When comparing insulin sensitive versus insulin-insensitive horses, 1 study64 concluded that the nuchal ligament adipose tissue contributed the most to the EMS proinflammatory profile, with an increased expression of IL-1β and IL6 in insulin-resistant horses despite no difference in other adipose tissue depots or systemic inflammatory mediators.

Differences in serum cytokine levels have also identified statistically significant differences between control and obese and/or EMS horses but with conflicting results. In a group of 110 light-breed horses, serum amyloid A concentrations were correlated with body condition score (BCS) and baseline insulin concentrations after correcting for age and sex, although correlations were not identified for TNFα or IL-6.66 Decreased concentrations of TNFα, IL-6, and IL-1 were identified in obese horses; however, endogenous cytokine levels were not associated with insulin levels.67 Serum IL-6 and activin A did positively correlate to insulin levels in another study68 but did not differ between EMS and control horses. These findings contrasted with several studies that identified (i) higher concentrations of TNFα in previously laminitic ponies versus nonlaminitic ponies;69 (ii) higher concentrations of TNFα and IL-6 in a group of obese, hyperinsulinemic ponies versus obese ponies;59 and (iii) an inverse association between TNFα and insulin sensitivity in 60 mares after adjusting for BCS and percent fat.70 Interestingly, neutrophil oxidative burst activity was found to be markedly increased in obese, hyperinsulinemic horses. However, in the same study67 peripheral cytokine gene expression was lower for IL-1 and IL-6 and there was no difference between groups in cytokine response after an inflammatory challenge, potentially reflecting a lack of a direct inflammatory response due to obesity and hyperinsulinemia in horses. However, EMS horses that had an oral sugar test performed with stevioside had an exaggerated inflammatory response systemically as compared to metabolically normal controls.71 Thus, the role of adipose tissue and inflammatory mediators in ID is still under investigation, but the presence of low-grade systemic inflammation could contribute to a metabolic OA phenotype, reproductive disease, and cardiovascular disease.

Myokines and adipokines

The roles of myokines and adipokines in horses are in the early stages of understanding, with an imbalance between the 2 being investigated as a contributing cause of metabolic syndrome. Myokines can enhance insulin sensitivity and myostatin inhibition also reduces inflammatory cytokines in muscle and adipose, while stimulating fatty acid oxidation.72 Local myostatin inhibitors can also result in increased skeletal muscle glucose dispersal, ameliorating insulin resistance while increasing growth rate and lean muscle mass,73 and a single nucleotide polymorphism (SNP) intron 2 of myostatin (MSTN)74,75 has been implicated as a powerful predictor of racing performance. This intronic SNP has been shown to be a marker of the true functional variant in a short interspersed element in the MSTN promoter that disrupts MSTN gene expression.76 Decreased MSTN gene expression has also been linked to increased insulin sensitivity in bulls, which lends support to this occurring in horses as well.77

Studies50,7881 have demonstrated that low serum high-molecular-weight (HMW) adiponectin is a consistent feature of the EMS phenotype, with adiponectin concentrations being inversely proportional to body mass. Adiponectin is considered insulin sensitizing, which makes it a biomarker of interest for a possible screening test for EMS versus leptin, which is more a marker of obesity in the horse.82 Adiponectin is released from adipose tissue in both low-molecular-weight trimers and HMW multimers. There is also a globular proteolytic fraction of the protein.83 The HMW form of adiponectin is considered the most metabolically active with the overall decrease in adiponectin concentration seen in obese human patients due to a decrease of the HMW form.84 Low levels of adiponectin have been associated with ID in the horse82 and are considered predictors of future laminitic events in ponies.85 The globular form of adiponectin also promotes glucose uptake and lipid oxidation while enhancing insulin signaling and myogenesis in humans but has an unknown role in the equine.83

Endocrine Effects on Musculoskeletal Health

Overtraining and muscle atrophy

Muscle itself is an endocrine organ, as addressed above with its production of myokines, and it plays a role in the athletic concern of overtraining. This condition is the result of a combination of excessive training and poor recovery. Clinical signs in the horse include decreased performance, weight loss, and behavior changes.8688 Possible endocrine-related causes behind this condition could include chronic glycogen depletion and increased reactive oxygen species and thus inflammatory pathways,89 and in both humans and horses, a dysregulation of the hypothalamus-pituitary-adrenal axis.87 As basal testing is not informative, stimulation tests of growth hormone and adrenocorticotropic hormone appear to offer the most promise as diagnostic tests for this condition, as humans affected by overtraining showed blunted responses.90

Muscle atrophy is classically associated with PPID in horses,6 but systemic inflammation in humans with MetS is thought to also result in sarcopenia.91 Past work in horses affected by EMS that evaluated selected inflammatory markers within the muscle did not find signs of inflammation present, but a more extensive evaluation of skeletal muscle of ID versus metabolically healthy horses would be beneficial.92 Of clinical interest, pergolide treatment by itself did not appear to ameliorate muscle atrophy in PPID-affected horses, suggesting that other muscle-building exercises, such as dynamic mobilization exercises, are needed.6,93

Lameness and obesity

Obesity is a common feature of EMS8 and has links to lameness, as heavier horses were more likely to not complete an endurance race due to lameness.94 In horses fed a high-energy diet to induce obesity, more gait asymmetry was seen and blinded judges gave them reduced performance scores.95 Obese humans have several reported negative effects on gait biomechanics such as development of angular limb deformities and increased joint stress and pain,96,97 and weight loss by itself has been shown to improve gait kinematics and alleviate pain.98 Humans with abdominal obesity (a component of MetS) and dynapenia (age-associated loss of muscle strength) have a faster decline in walking speed with age than healthy controls,99 which may have ramifications on performance if this holds true in our older equine athletes. Another musculoskeletal condition that has links to endocrine disease is osteochondrosis dessicans (OCD). Increased frequency of OCD and insulin resistance was seen in foals and yearlings born of obese mares, which is discussed further below (see endocrine effects on reproductive health).9 Readers interested in further details on specific equine performance conditions such as laminitis (a critically important and commonly recognized sequela of equine metabolic disorders) are invited to read the companion Currents in One Health by Manfredi et al, JAVMA, February 2023. Endocrine disorders in humans have also been associated with a metabolic OA subtype,100 as well as tendon and ligament disease, for which we will now go into more detail.100108

Metabolic osteoarthritis

Previous research efforts have indicated the appropriateness of using the equine as a model for human OA because the forces exerted with exercise on a human joint are similar to those observed in the horse, so it is reasonable to take a one-health approach considering shared pathophysiology.109111 Type II diabetes, which can be associated with MetS, has been linked to development of severe OA in humans.107 Furthermore, individuals with OA of which metabolic OA is now considered a subtype,2 have annual healthcare costs ∼$11,000 greater than non-OA-affected individuals.112 The precise role that MetS plays in the development of metabolic OA has yet to be fully elucidated but could be involved via several mechanisms.104 One possibility is that the hyperglycemia and low-grade systemic inflammation that occurs during MetS can diffuse into the joint from systemic circulation creating a toxic environment.2,113 Dyslipidemia could also result in ectopic lipid deposition in the chondrocytes, and hypertension could cause ischemia of the subchondral bone.2 While previous work was not able to distinguish between obesity or MetS as the cause of the OA, recent work has shown that MetS by itself can contribute to the development of excessive knee OA pain, with studies100,101 suggesting that gut dysbiosis may play a role in this syndrome. The idea that gut dysbiosis can contribute to joint pain is supported by the fact that oral resveratrol supplementation has been shown to alleviate equine tarsal OA pain and 114 improve insulin regulation,30,115 while also having a beneficial effect on the gut microbiome in rat and mice models of MetS.116,117 Given that obesity has been reported in 30 to 45% of horses118,119 and that some riding disciplines seem to favor the overweight phenotype (dressage, hunters, ponies), there is a concern that metabolic OA could be more of a present and future problem in horses than is currently recognized.120

Biomarkers for metabolic OA

In humans, the adipokines adiponectin, leptin, and resistin have been found to be potential biomarkers of metabolic OA.108 Systemically, adiponectin has contradicting information about its role, as some studies105,121 found that it causes metabolic OA while others claim it could have a protective effect. Another study122 demonstrated that increased levels of leptin can contribute to metabolic OA systemically. While there are currently no studies in horses looking at the possibility of adipokines as synovial fluid biomarkers in metabolic horses with OA, 2 human studies106,123 looked at adipokine expression in synovial fluid and serum that have OA in patients with and without MetS. Both studies106,123 found that humans with metabolic OA had higher serum leptin concentrations and lower serum adiponectin concentrations compared to humans without metabolic OA. The studies106 also looked at concentrations in the synovial fluid and the protein expression of the infrapatellar fat pad and found greater expression and concentrations of leptin and less expression and concentrations of adiponectin in humans with metabolic OA. These findings support leptin and adiponectin as both biomarkers for and possible therapeutic targets in patients with metabolic OA. Another study106 evaluated resistin concentrations in serum or synovial fluid and did not find a difference between MetS or non-MetS patients; however, there was a positive correlation between resistin serum and synovial fluid concentrations and OA grade, as well as a negative correlation between adiponectin serum and synovial fluid concentrations and OA grade. In horses with ID, resistin was elevated in plasma, which was attributed to the inflammatory nature of severe ID, and evaluation of its presence in equine synovial fluid and synovium should be examined.124 While other traditional biomarkers for OA should be evaluated in ID and metabolically healthy horses,109,125,126 future research for novel biomarkers should also include assessment with the multiomic approaches as discussed below.127

Tendon and ligament injury

Individuals with type II diabetes have a 3 times higher risk of tendon injury or rupture than control individuals, which can be due to the weight of the individual, inflammatory cytokines, or hyperglycemia and hypercholesterolemia with subsequent production of advanced glycation end-products.128 EMS has not been identified as a risk factor for tendon and ligament injury in horses, but given the findings in humans, more research should be pursued. However, PPID in horses has been identified as a risk factor for suspensory ligament degeneration, potentially due to excess circulating cortisol.5,129 This is similar in humans where Cushing’s syndrome has been implicated in Achilles tendon rupture.130,131

Exercise to mitigate the effects of endocrine disease

In humans, there is some indication that an active lifestyle and diet modification have beneficial effects on prevention of MetS and can relieve the symptoms associated with OA.132,133 Similar findings have been seen in horses, and exercise and dietary changes are the primary management strategies employed when treating animals with EMS or ID.134136 A novel type of exercise investigated in humans, which has been gaining popularity in horses, that could be an effective therapeutic intervention, is resistance band training. In recent studies,137,138 resistance band training and a restricted calorie diet ameliorated many of the symptoms of MetS, as did just 1 hour a day of resistance band training alone. Resistance band training in the horse has been shown to increase thoracolumbar stability,139 although it was not shown to activate and strengthen the multifidus muscle while walking over cavalettis.140 Resistance band training is attractive as a therapy as it can be performed at the walk, allowing previously laminitic ponies and horses to still exercise. More work should be done to concurrently assess endocrine status changes and muscle development with resistance band training.

Endocrine Effects on Cardiovascular Health

Football linemen, while athletes, have higher percentages of cardiovascular death and MetS than nonlinemen and the general public.141 The effects of EMS on cardiovascular health have received less attention, as cardiovascular disease-related death is uncommon in equines in general. However, EMS horses during a euglycemic-hyperinsulinemic clamp procedure demonstrated maintenance of systolic, diastolic, and mean arterial pressure over time as compared to control horses where blood pressure decreased, suggesting cardiac dysfunction in the EMS group.142 Ponies with EMS have also been found to have myocardial hypertrophy correlated to insulin responses during dynamic testing as compared to controls but no difference in blood pressure.10 Shetland ponies, a high-risk breed for EMS, when placed on a high-energy diet, developed increased blood pressure and ultimately cardiac hypertrophy, albeit no arrhythmias, when compared to ponies on a control diet. This suggests that our equine athletes that are considered high-risk breeds should not be fed the high-energy diets that are often given to performance horses. Performance horses with EMS should also have more attention paid to clinical signs that could indicate early cardiac dysfunction.

Endocrine Effects on Reproductive Health

Reproductive health

Obesity and MetS are linked to infertility in humans.143,144 In mares with EMS, the intrafollicular environment, ie, the follicle, the oocyte, and the uterus, is affected, and in the latter case inflamed, which could have an impact on breeding success.12,145,146 Other work has shown the negative impact of endocrine disease (both EMS and PPID) on reproductive seasonality and ovulation, with subsequent increased incidences of pregnancy complications.147 Selection of desirable traits for an equine athlete during breeding comes with both risk and reward; however, optimization of whole body health is important for short-term and long-term breeding and athletic success. There are also costs (more time away from sport, financial ramifications) associated with a missed breeding season due to an endocrine disorder. Athletic mares with endocrine disease that are having issues with infertility or maintaining a pregnancy may require more aggressive therapeutic management and possibly need assisted reproduction techniques such as embryo transfer.

Developmental origin of metabolic disease and placental function

The concept that the maternal environment can lead to permanent fetal adaptions that manifest at different stages of the offspring’s life is a well-recognized theory known as the Developmental Origin of Health and Disease.148 Data in the horse are relatively scarce but have highlighted the role of the maternal environment on postnatal body conformation, sexual maturity, immune response, metabolism, osteoarticular state, and cardiovascular function of the foal.149,150 A specific area of interest has been the evaluation of the effect of maternal overnutrition and obesity on foal outcomes. Overfeeding of mares during pregnancy to induce late-gestation obesity resulted in foals with delayed testicular maturation and insulin resistance up to 2 years of age.151 Maternal overnutrition was also found to reduce colostrum IgG concentrations, resulting in lower foal serum IgG levels at 12 and 18 hours of age, although none of the foals were diagnosed with failure of passive transfer.152 Further, foals born to mares that were obese at the time of insemination and throughout their pregnancy had higher serum amyloid A concentrations, insulin resistance at 6 and 18 months of age, and an increased incidence of OCD at 1 year of age.94 Embryos from obese mares had increased expression of inflammatory genes, lipid homeostasis, and oxidative stress.146 These studies indicate that maternal obesity can have a deleterious affect on offspring health, reproduction, metabolism, and athletic performance. Importantly, although the obese mares present in these studies had metabolic values higher than nonobese mares, values were not consistent with a diagnosis of EMS or impaired insulin sensitivity only occurred at certain points during gestation. Therefore, the effect of EMS on foal health remains an area to be investigated.

The link between maternal ID and adverse health outcomes in their offspring is well established in diabetic pregnant women and has been linked to placental pathology. Increased placental weight and thickness have been correlated with higher fetal birth weights and shoulder dystocia, doubling the risk of a nonelective cesarean section in diabetic women.153 In addition, placental histological changes include an increased incidence of ischemia, infarction, fibrinoid necrosis, villous immaturity, edema, choriangiosis, and fetal vessel thrombosis.154 These lesions result in altered fetoplacental circulation and impaired oxygen transport and have been independently associated with stillbirths and fetal morbidity,154 confirming significant placental pathology in women with gestational diabetes.

Compared to other species, the equine fetus relies more heavily on placental transfer of glucose and nutrients due to the limited degree of deamination and gluconeogenesis,155 which could indicate an even more critical role of the placenta in EMS-induced fetal pathologies. In addition, this early fetal programming may lead to long-term insulin resistance in the offspring, increasing the risk of juvenile and adult obesity and EMS as seen in humans.156 Obesity has been shown to limit a horse’s athletic career through a reduced physiological exercise response and athletic performance and increased joint strain in adults, as well as having a higher incidence of developmental orthopedic problems in growing horses.95 Further, having an increased risk of EMS represents a vicious cycle that will continue to perpetuate this disease, increasing its economic impact on the industry.

Multiomic Insights into Pathophysiology, Novel Diagnostic Tests, and Biomarkers of Disease

Genomics

The primary goal of genetic discovery is to gain insight into the pathogenesis of a trait for the development of genetic tests, which can be used to identify affected individuals and to help guide breeding decisions. This is of particular importance for EMS and PPID as the current diagnostic tests are imperfect and often miss early onset individuals. Therefore, there is a need to be able to identify high-risk individuals before they develop clinical signs to implement proper environmental or pharmaceutical intervention thus securing longer athletic life spans while improving horse welfare.

Unlike Mendelian (simple) traits where a single genetic mutation leads to a phenotype, endocrine disorders are often complex genetic diseases with dozens to hundreds of genetic risk alleles contributing to their phenotypic expression. Further, these genetic risk alleles can be unique to specific groups or have variable allele frequencies, variant effect size (amount of phenotypic variation explained by the variant), and penetrance across groups. Therefore, a single genetic test would provide minimal information about an individual’s genetic risk for an endocrine disorder and instead requires a panel of well-validated shared and group-specific genetic risk alleles in conjunction with their variant effect size (Table 1). Interpretation of these results would not be as straightforward as Mendelian diseases and would benefit from the consultation of a geneticist or genetic counselor, particularly when including environmental risk factors to determine an individual’s overall risk of developing disease or when evaluating the impact of specific genetic alleles on the panel that have been associated with muscle or reduced athletic function.

Table 1

Hypothetical example for an EMS genetic risk panel.

Horse 1 Horse 2
Genetic Risk Breed Affect Size Allele relative risk Pony Morgan
Allele 1 All Large 0.7 NEG NEG
Allele 2 All Large 0.6 POS NEG
Allele 6 All Small 0.2 POS POS
Allele 4 Morgan Moderate 0.5 NEG POS
Allele 5 Morgan Moderate 0.5 NEG POS
Arabians
Allele 7 Ponies Large 0.9 POS NEG
Allele ….
Genetic RR 1.7 1.2

A genetic panel that will accurately assess genetic risk for a complex genetic disease would need to include both shared and breed specific risk alleles and their estimated allelic affect size and relative risk. For example, if the genotypes for a hypothetical pony (horse 1) include 2 across-breed alleles of large and small affect size and 1 pony-specific allele of large effect, then that individual would have a hypothetical EMS genetic relative risk of 1.7. If we compare that to the genotypes for a hypothetical Morgan horse (horse 2) who has 1 across breed allele of small effect and 2 breed specific risk alleles of moderate affect, then this individual’s hypothetical EMS genetic relative risk would be less than horse 1.

NEG = Negative. POS = Positive. RR = Relative risk. (...) = indicates the continuation of the number of alleles that would be on the panel since it will be more than seven.

Toward these goals, advances have been made in the genetic discovery of EMS. There is concrete evidence that EMS has a strong genetic contribution with hundreds of shared and breed-specific regions of the genome contributing to its development.157,158 However, despite the identification of multiple associated genomic regions, only 2 genetic variants have been proposed,158,159 1 of which was not replicated in follow-up studies.160,161 Therefore, it is imperative to continue to explore the specific genes and genetic risk factors for EMS. However, given the complexity of the EMS phenotype and the high percentage of associated SNPs located in noncoding regions of the genome,162 an integrated multiomics approach (Figure 2) must be implemented to aid in this genetic discovery. Most previous studies of EMS have focused on the individual effects of the genome,157,158,162 transcriptome,163 or metabolome,164,165 and while each type of omics data provided a list of differences associated with EMS, it only represented 1 layer of the genetic effect that limited the identification of specific risk alleles. In contrast, the rationale for a multiomics approach is that phenotypic effects may be expressed through alterations in gene expression or regulation, or protein abundance, function, or regulation, which manifest through alterations in metabolic pathways and functions, aiding in the genetic discovery of causative alleles. This approach, used successfully for genomic discovery in humans with complex genetic diseases, identified four associated genomic regions for EMS in Arabians166 and was considered a priority in the USDA Blueprint for Animal Genome Research toward bringing genome to phenome.167

Of particular interest is the inclusion of the epigenome, a layer of omics data that has yet to be explored in horses with EMS. Epigenetics are cell-specific changes in gene expression that are independent of DNA sequences. Epigenetic modifications are initially established in utero and can be affected by maternal health and nutrition; however, many epigenetic changes can be altered throughout the offspring’s life secondary to environmental exposures. In humans, a link between gestational diabetes and risk of metabolic disorders in their offspring has been well established, and these changes have been associated with alterations in the placental or offspring epigenome.168 Another study169 identified differential DNA methylation levels near the leptin genes, which were found to mediate the effects of maternal blood glucose concentrations on neonatal leptin concentrations. Therefore, the effect of the mare’s EMS status may have a significant impact on her offspring’s risk of ID and warrants further exploration.

The contribution of genetics to PPID has not yet been proven. Therefore, the first step is to confirm a genetic etiology, which will require using big data in well-phenotyped individuals. Whole-genome sequencing or SNP data could be used to estimate the heritability of PPID and to perform a genome-wide association analysis for the identification of putative functional alleles. Alternatively, a candidate gene approach would determine if horses with PPID suffer from the same causal variants as Parkinson disease, a human disease also caused by dopaminergic neurodegeneration in aged individuals. This approach would be particularly useful in horses with suspected familial PPID (high prevalence of PPID within a line) as these cases are more likely to contain few genes of a larger impact compared to sporadic cases. Finally, a multiomics approach could identify biological pathways responsible for PPID. Thus, these genetic discoveries will be essential to help elucidate the underlying pathophysiology of PPID and to determine an appropriate genetic test to accurately identify high-risk individuals.

Metabolomics

Metabolomics, the study of molecules involved in cellular metabolism, refers to the global interrogation of the biochemical components in a biological sample such as serum, plasma, urine, feces, saliva, or cerebrospinal fluid. Small molecule metabolites such as lipids, amino acids, peptides, nucleic acids, organic acids, fatty acids, vitamins, carbohydrates, hormones, and steroids are the end products of cellular regulatory processes. As the ultimate response of biological systems to genetic or environmental changes, the comprehensive measurement of metabolites reflects perturbations in metabolism, thus providing insight into biological mechanisms and pathogenesis of disease through an understanding of molecular pathways. In addition, metabolomics defines metabolic changes in both the physiological and pathological states. On the most basic level, metabolomics can differentiate between healthy individuals and diseased individuals.170,171

Studies172175 in humans have identified characteristic metabolic signatures of type II diabetes mellitus, obesity, and fatty liver disease. A consistent pattern of reduced glycine and acylcarnitines176178 with increased concentrations of valine, isoleucine, and α-hydroxybuterate179,180 has been associated with both basal and dynamic measures of insulin resistance.177,178 The metabolic perturbations that characterize and contribute to type II diabetes mellitus are evident years before disease onset, and comprehensive metabolomic profiles have elucidated alterations in novel metabolic pathways implicated in disease development.

In horses, examination of the serum metabolome in a cohort of Welsh ponies demonstrated significant differences in metabolites involved in the tricarboxylic acid (TCA) cycle, fatty acid metabolism, and branched-chain amino acid metabolism when comparing animals with and without the EMS phenotype.165 In addition, evaluation of the fecal metabolome of obese horses and nonobese horses noted significant differences in metabolites of the TCA.181

Metabolomics in human sport and exercise has revealed insights into muscle energetics, and it is just starting to be of interest in the horse.182 In a paper183 on acute fatigue and training in normal horses, increases in lipids were noted in response to training. This increased abundance of lipids, if present after training in horses with ID and lipid dysregulation, could be of concern, especially as this response was seen in humans with MetS after high-intensity interval training.184 In that study,183 the lipid increase was more pronounced after afternoon training sessions. In a second study,185 humans with MetS saw greater improvements in insulin sensitivity after a morning workout session. If these findings hold true for ID horses, morning training sessions may be the best time to perform exercise for our EMS-affected animals. Used as a diagnostic tool, metabolomics may aid in early detection of disease and ID before development of laminitis,164 which allows for earlier intervention into prevention and treatment, while as shown with the human exercise work, it may guide other management strategies.

Transcriptomics

Transcriptomics, or analysis of RNA transcripts made by a genotype at a certain time, promises to be a powerful way to assess genes that affect both metabolic health and performance.186,187 In Arabians, a breed “at risk” for EMS, there was an upregulation in the PI3K-Akt signaling pathways in trained endurance horses as compared to untrained horses, suggesting a more efficient glucose uptake, which can explain the insulin-sensitizing effect of exercise.188 Transcriptomic analysis also has the ability to analyze gene coexpression networks and reveal their biological functions, in different tissues from the same individuals, assessing “cross-talk” as has been described in muscle and adipose tissue in horses.163 A human paper189 taking this approach compared metabolically healthy and unhealthy obese individuals and found IL-6 and IL-1β were differentially coexpressed genes, and similar studies could better reveal the pathophysiology of endocrine disorders in the horse.

Proteomics

Proteomics are also an up-and-coming research tool to assess therapeutic interventions and explore pathophysiology of disease. In a paper190 investigating dietary omega-3 supplementation in ID mares, proteomics of the uterine fluid found inflammatory biomarkers decreased when compared to controls and ID mares not receiving the supplement. Proteomic approaches have also shown utility in assessing treatment and biomarkers for equine muscle diseases191 and biomarkers for human OA.192 In humans with type II diabetes mellitus, GLUT4 protein expression increased with exercise, helping to increase their insulin sensitivity.193

Microbiomics

The gastrointestinal tract of humans and animals is composed of numerous microorganisms that play a role in overall health. In humans, the gastrointestinal microbiome has been shown to be an important factor in the development of obesity and type II diabetes and it is postulated to play a role in the development of metabolic OA.194,195 The complex microbial population of the equine intestinal tract plays a key role in health and disease, and the diverse, variable composition and complexity of this population are starting to be revealed, now that methods to characterize the genetic makeup of the microbiota have been developed.196199 In a small cohort of horses with EMS, there was a decrease in microbiota diversity compared to healthy controls.200 In addition, horses with chronic laminitis were noted to have differences in microbiota diversity compared to healthy controls. In mildly ID ponies, the introduction of pasture was able to perturb their microbiome more significantly than metabolically healthy ponies.26 In addition, ID horses on metformin, a common treatment for EMS, experienced a microbiota change similar to that seen in humans on that drug.32 The availability of next-generation sequencing and bioinformatics methods offers an opportunity to improve understanding of the role of the gastrointestinal microbiome in the pathogenesis of metabolic disorders and ID.

In summary, the effects of endocrine disease on musculoskeletal, cardiovascular, and reproductive health have repercussions on athletic performance in both humans and horses. Multiomic approaches provide information about molecular and cellular processes in endogenous and exogenous pathways and insight into physiology and pathophysiology of an individual. Further, these approaches allow for better connection of genotype and phenotype leading to identification of biomarkers that can provide insight for novel diagnostic and therapeutic targets and strategies. Formulation of a management plan for individuals with metabolic disorders should focus on interventions that improve overall health and athletic performance.

Acknowledgments

None of the authors have any financial or personal relationships that could bias the content of the manuscript.

References

  • 1.

    Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287(3):356359.

    • Search Google Scholar
    • Export Citation
  • 2.

    Zhuo Q, Yang W, Chen J, Wang Y. Metabolic syndrome meets osteoarthritis. Nat Rev Rheumatol. 2012;8(12):729737. doi:10.1038/nrrheum.2012.135

    • Search Google Scholar
    • Export Citation
  • 3.

    Monira Hussain S, Wang Y, Cicuttini FM, et al. Incidence of total knee and hip replacement for osteoarthritis in relation to the metabolic syndrome and its components: a prospective cohort study. Semin Arthritis Rheum. 2014;43(4):429436. doi:10.1016/j.semarthrit.2013.07.013

    • Search Google Scholar
    • Export Citation
  • 4.

    Morgan R, Keen J, McGowan C. Equine metabolic syndrome. Vet Rec. 2015;177(7):173179. doi:10.1136/vr.103226

  • 5.

    Hofberger S, Gauff F, Licka T. Suspensory ligament degeneration associated with pituitary pars intermedia dysfunction in horses. Vet J. 2015;203(3):348350. doi:10.1016/j.tvjl.2014.12.037

    • Search Google Scholar
    • Export Citation
  • 6.

    Banse HE, Whitehead AE, McFarlane D, Chelikani PK. Markers of muscle atrophy and impact of treatment with pergolide in horses with pituitary pars intermedia dysfunction and muscle atrophy. Domest Anim Endocrinol. 2021;76:106620. doi:10.1016/j.domaniend.2021.106620

    • Search Google Scholar
    • Export Citation
  • 7.

    Geor R, Frank N. Metabolic syndrome-From human organ disease to laminar failure in equids. Vet Immunol Immunopathol. 2009;129(3–4):151154. doi:10.1016/j.vetimm.2008.11.012

    • Search Google Scholar
    • Export Citation
  • 8.

    Frank N, Geor RJ, Bailey SR, Durham AE, Johnson PJ. Equine metabolic syndrome. J Vet Intern Med. 2010;24(3):467475. doi:10.1111/j.1939-1676.2010.0503.x

    • Search Google Scholar
    • Export Citation
  • 9.

    Robles M, Nouveau E, Gautier C, et al. Maternal obesity increases insulin resistance, low-grade inflammation and osteochondrosis lesions in foals and yearlings until 18 months of age. PLoS One. 2018;13(1):e0190309. doi:10.1371/journal.pone.0190309

    • Search Google Scholar
    • Export Citation
  • 10.

    Heliczer N, Gerber V, Bruckmaier R, van der Kolk JH, de Solis CN. Cardiovascular findings in ponies with equine metabolic syndrome. J Am Vet Med Assoc.2017;250(9):10271035. doi:10.2460/javma.250.9.1027

    • Search Google Scholar
    • Export Citation
  • 11.

    D’Fonseca NMM, Beukers, M, Wijnberg ID, et al. Effect of a long-term high-energy diet on cardiovascular parameters in Shetland pony mares. J Vet Intern Med. 2021;35(5):24272436. doi:10.1111/jvim.16229

    • Search Google Scholar
    • Export Citation
  • 12.

    Sessions-Bresnahan DR, Carnevale EM. The effect of equine metabolic syndrome on the ovarian follicular environment. J Anim Sci. 2014;92(4):14851494. doi:10.2527/jas.2013-7275

    • Search Google Scholar
    • Export Citation
  • 13.

    Durham AE, Frank N, McGowan CM, et al. ECEIM consensus statement on equine metabolic syndrome. J Vet Intern Med. 2019;33(2):335349. doi:10.1111/jvim.15423

    • Search Google Scholar
    • Export Citation
  • 14.

    Wilcox G. Insulin and insulin resistance. Clin Biochem Rev. 2005;26(2):1939.

  • 15.

    Thorens B. GLUT2, glucose sensing and glucose homeostasis. Diabetologia. 2015;58(2):221232. doi:10.1007/s00125-014-3451-1

  • 16.

    McCulloch LJ, van de Bunt M, Braun M, Frayn KN, Clark A, Gloyn AL. GLUT2 (SLC2A2) is not the principal glucose transporter in human pancreatic beta cells: implications for understanding genetic association signals at this locus. Mol Genet Metab. 2011;104(4):648653. doi:10.1016/j.ymgme.2011.08.026

    • Search Google Scholar
    • Export Citation
  • 17.

    Bratanova-Tochkova TK, Cheng H, Daniel S, et al. Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes. 2002;51(suppl 1):S83S90. doi:10.2337/diabetes.51.2007.s83

    • Search Google Scholar
    • Export Citation
  • 18.

    Pratt SE, Geor RJ, McCutcheon LJ. Repeatability of 2 methods for assessment of insulin sensitivity and glucose dynamics in horses. J Vet Intern Med. 2005;19(6):883888.

    • Search Google Scholar
    • Export Citation
  • 19.

    Toth F, Frank N, Elliott SB, Perdue K, Geor RJ, Boston RC. Optimisation of the frequently sampled intravenous glucose tolerance test to reduce urinary glucose spilling in horses. Equine Vet J. 2009;41(9):844851. doi:10.2746/042516409x439661

    • Search Google Scholar
    • Export Citation
  • 20.

    Czech MP, Buxton JM. Insulin action on the internalization of the GLUT4 glucose transporter in isolated rat adipocytes. J Biol Chem. 1993;268(13):91879190.

    • Search Google Scholar
    • Export Citation
  • 21.

    de Graaf-Roelfsema E. Glucose homeostasis and the enteroinsular axis in the horse: a possible role in equine metabolic syndrome. Vet J. 2014;199(1):1118. doi:10.1016/j.tvjl.2013.09.064

    • Search Google Scholar
    • Export Citation
  • 22.

    Polonsky KS, Given BD, Hirsch L, et al. Quantitative study of insulin secretion and clearance in normal and obese subjects. J Clin Invest. 1988;81(2):435441. doi:10.1172/jci113338

    • Search Google Scholar
    • Export Citation
  • 23.

    Tóth F, Frank N, Martin-Jiménez T, Elliott SB, Geor RJ, Boston RC. Measurement of C-peptide concentrations and responses to somatostatin, glucose infusion, and insulin resistance in horses. Equine Vet J. 2010;42(2):149155. doi:10.2746/042516409x478497

    • Search Google Scholar
    • Export Citation
  • 24.

    Faber OK, Christensen K, Kehlet H, Madsbad S, Binder C. Decreased insulin removal contributes to hyperinsulinemia in obesity. J Clin Endocrinol Metab. 1981;53(3):618621. doi:10.1210/jcem-53-3-618

    • Search Google Scholar
    • Export Citation
  • 25.

    Bamford NJ, Baskerville CL, Harris PA, Bailey SR. Postprandial glucose, insulin, and glucagon-like peptide-1 responses of different equine breeds adapted to meals containing micronized maize. J Anim Sci. 2015;93(7):33773383. doi:10.2527/jas.2014-8736

    • Search Google Scholar
    • Export Citation
  • 26.

    Fitzgerald DM, Walsh DM, Sillence MN, Pollitt CC, de Laat MA. Insulin and incretin responses to grazing in insulin-dysregulated and healthy ponies. J Vet Intern Med. 2019;33(1):225232. doi:10.1111/jvim.15363

    • Search Google Scholar
    • Export Citation
  • 27.

    Frank N, Walsh DM. Repeatability of oral sugar test results, glucagon-like peptide-1 measurements, and serum high-molecular-weight adiponectin concentrations in horses. J Vet Intern Med. 2017;31(4):11781187. doi:10.1111/jvim.14725

    • Search Google Scholar
    • Export Citation
  • 28.

    Nauck MA, Quast DR, Wefers J, Meier JJ. GLP-1 receptor agonists in the treatment of type 2 diabetes–state-of-the-art. Mol Metab. 2021;46:101102. doi:10.1016/j.molmet.2020.101102

    • Search Google Scholar
    • Export Citation
  • 29.

    Stefanovski D, Robinson MA, Van Eps A. Effect of a GLP-1 mimetic on the insulin response to oral sugar testing in horses. BMC Vet Res. 2022;18(1):294. doi:10.1186/s12917-022-03394-2

    • Search Google Scholar
    • Export Citation
  • 30.

    Manfredi JM, Stapley ED, Nadeau JA, Nash D. Investigation of the effects of a dietary supplement on insulin and adipokine concentrations in equine metabolic syndrome/insulin dysregulation. J Equine Vet Sci. 2020;88:102930. doi:10.1016/j.jevs.2020.102930

    • Search Google Scholar
    • Export Citation
  • 31.

    Durham AE. Therapeutics for equine endocrine disorders. Vet Clin North Am Equine Pract. 2017;33(1):127139. doi:10.1016/j.cveq.2016.11.003

    • Search Google Scholar
    • Export Citation
  • 32.

    Ericsson AC, Johnson PJ, Gieche LM, et al. The influence of diet change and oral metformin on blood glucose regulation and the fecal microbiota of healthy horses. Animals (Basel). 2021;11(4):976. doi:10.3390/ani11040976

    • Search Google Scholar
    • Export Citation
  • 33.

    Hustace JL, Firshman AM, Mata JE. Pharmacokinetics and bioavailability of metformin in horses. Am J Vet Res. 2009;70(5):665668. doi:10.2460/ajvr.70.5.665

    • Search Google Scholar
    • Export Citation
  • 34.

    Rendle DI, Rutledge F, Hughes KJ, Heller J, Durham AE. Effects of metformin hydrochloride on blood glucose and insulin responses to oral dextrose in horses. Equine Vet J. 2013;45(6):751754. doi:10.1111/evj.12068

    • Search Google Scholar
    • Export Citation
  • 35.

    Tinworth KD, Boston RC, Harris PA, Sillence MN, Raidal SL, Noble GK. The effect of oral metformin on insulin sensitivity in insulin-resistant ponies. Vet J. 2012;191(1):7984. doi:10.1016/j.tvjl.2011.01.015

    • Search Google Scholar
    • Export Citation
  • 36.

    Frank N, Elliott SB, Boston RC. Effects of long-term oral administration of levothyroxine sodium on glucose dynamics in healthy adult horses. Am J Vet Res. 2008;69(1):7681. doi:10.2460/ajvr.69.1.76

    • Search Google Scholar
    • Export Citation
  • 37.

    Geor RJ, Harris P. Dietary management of obesity and insulin resistance: countering risk for laminitis. Vet Clin North Am Equine Pract. 2009;25(1):5165. doi:10.1016/j.cveq.2009.02.001

    • Search Google Scholar
    • Export Citation
  • 38.

    Gong IY, Atzema CL, Lega IC, et al. Levothyroxine dose and risk of atrial fibrillation: a nested case-control study. Am Heart J. 2021;232:4756. doi:10.1016/j.ahj.2020.09.016

    • Search Google Scholar
    • Export Citation
  • 39.

    Kritchevsky J, Olave C, Tinkler S, et al. A randomised, controlled trial to determine the effect of levothyroxine on Standardbred racehorses. Equine Vet J. 2022;54(3):584591. doi:10.1111/evj.13480

    • Search Google Scholar
    • Export Citation
  • 40.

    Delahanty LM, Pan Q, Jablonski KA, et al. Genetic predictors of weight loss and weight regain after intensive lifestyle modification, metformin treatment, or standard care in the Diabetes Prevention Program. Diabetes Care. 2012;35(2):363366. doi:10.2337/dc11-1328

    • Search Google Scholar
    • Export Citation
  • 41.

    Brasnyo P, Molnar GA, Mohas M, et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr. 2011;106(3):383389. doi:10.1017/s0007114511000316

    • Search Google Scholar
    • Export Citation
  • 42.

    Kellon EM, Gustafson KM. Use of the SGLT2 inhibitor canagliflozin for control of refractory equine hyperinsulinemia and laminitis. Open Vet J. 2022;12(4):511518. doi:10.5455/OVJ.2022.v12.i4.14

    • Search Google Scholar
    • Export Citation
  • 43.

    Sundra T, Kelty E, Rendle D. Preliminary observations on the use of ertugliflozin in the management of hyperinsulinemia and laminitis in 51 horses: a case series. Equine Vet Educ. 2022;00:110. doi:10.1111/eve.13738

    • Search Google Scholar
    • Export Citation
  • 44.

    Engin A. The definition and prevalence of obesity and metabolic syndrome. Adv Exp Med Biol. 2017;960:117. doi:10.1007/978-3-319-48382-5_1

    • Search Google Scholar
    • Export Citation
  • 45.

    Perseghin G. Muscle lipid metabolism in the metabolic syndrome. Curr Opin Lipidol. 2005;16(4):416420. doi:10.1097/01.mol.0000174401.07056.56

    • Search Google Scholar
    • Export Citation
  • 46.

    Arner P. Insulin resistance in type 2 diabetes: role of fatty acids. Diabetes Metab Res Rev. 2002;18(suppl 2):S5S9. doi:10.1002/dmrr.254

    • Search Google Scholar
    • Export Citation
  • 47.

    Treiber KH, Kronfeld DS, Hess TM, Byrd BM, Splan RK, Staniar WB. Evaluation of genetic and metabolic predispositions and nutritional risk factors for pasture-associated laminitis in ponies. J Am Vet Med Assoc. 2006;228(10):15381545. doi:10.2460/javma.228.10.1538

    • Search Google Scholar
    • Export Citation
  • 48.

    Carter RA, Treiber KH, Geor RJ, Douglass L, Harris PA. Prediction of incipient pasture-associated laminitis from hyperinsulinaemia, hyperleptinaemia and generalised and localised obesity in a cohort of ponies. Equine Vet J. 2009;41(2):171178.

    • Search Google Scholar
    • Export Citation
  • 49.

    Bailey SR, Habershon-Butcher JL, Ransom KJ, Elliott J, Menzies-Gow NJ. Hypertension and insulin resistance in a mixed-breed population of ponies predisposed to laminitis. Am J Vet Res. 2008;69(1):122129. doi:10.2460/ajvr.69.1.122

    • Search Google Scholar
    • Export Citation
  • 50.

    Frank N, Elliott SB, Brandt LE, Keisler DH. Physical characteristics, blood hormone concentrations, and plasma lipid concentrations in obese horses with insulin resistance. J Am Vet Med Assoc. 2006;228(9):13831390. doi:10.2460/javma.228.9.1383

    • Search Google Scholar
    • Export Citation
  • 51.

    Coleman MC, Walzem RL, Kieffer AJ, Minamoto T, Suchodolski J, Cohen ND. Novel lipoprotein density profiling in laminitic, obese, and healthy horses. Domest Anim Endocrinol. 2019;68:9299. doi:10.1016/j.domaniend.2018.11.003

    • Search Google Scholar
    • Export Citation
  • 52.

    Sethi JK, Vidal-Puig AJ. Thematic review series: adipocyte biology. Adipose tissue function and plasticity orchestrate nutritional adaptation. J Lipid Res. 2007;48(6):12531262. doi:10.1194/jlr.R700005-JLR200

    • Search Google Scholar
    • Export Citation
  • 53.

    Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46(11):23472355. doi:10.1194/jlr.M500294-JLR200

    • Search Google Scholar
    • Export Citation
  • 54.

    Barchetta I, Cimini FA, Ciccarelli G, Baroni MG, Cavallo MG. Sick fat: the good and the bad of old and new circulating markers of adipose tissue inflammation. J Endocrinol Invest. 2019;42(11):12571272. doi:10.1007/s40618-019-01052-3

    • Search Google Scholar
    • Export Citation
  • 55.

    Arner E, Westermark PO, Spalding KL, et al. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes. 2010;59(1):105109. doi:10.2337/db09-0942

    • Search Google Scholar
    • Export Citation
  • 56.

    De Lucia Rolfe E, Ong KK, Sleigh A, Dunger DB, Norris SA. Abdominal fat depots associated with insulin resistance and metabolic syndrome risk factors in black African young adults. BMC Public Health. 2015;15:1013. doi:10.1186/s12889-015-2147-x

    • Search Google Scholar
    • Export Citation
  • 57.

    Grandl G, Wolfrum C. Hemostasis, endothelial stress, inflammation, and the metabolic syndrome. Semin Immunopathol. 2018;40(2):215224. doi:10.1007/s00281-017-0666-5

    • Search Google Scholar
    • Export Citation
  • 58.

    Henninger AM, Eliasson B, Jenndahl LE, Hammarstedt A. Adipocyte hypertrophy, inflammation and fibrosis characterize subcutaneous adipose tissue of healthy, non-obese subjects predisposed to type 2 diabetes. PLoS One. 2014;9(8):e105262. doi:10.1371/journal.pone.0105262

    • Search Google Scholar
    • Export Citation
  • 59.

    Basinska K, Marycz K, Sieszek A, Nicpon J. The production and distribution of IL-6 and TNF-a in subcutaneous adipose tissue and their correlation with serum concentrations in Welsh ponies with equine metabolic syndrome. J Vet Sci. 2015;16(1):113120. doi:10.4142/jvs.2015.16.1.113

    • Search Google Scholar
    • Export Citation
  • 60.

    Reynolds A, Keen JA, Fordham T, Morgan RA. Adipose tissue dysfunction in obese horses with equine metabolic syndrome. Equine Vet J. 2019;51(6):760766. doi:10.1111/evj.13097

    • Search Google Scholar
    • Export Citation
  • 61.

    Das S, Das DK. Anti-inflammatory responses of resveratrol. Inflamm Allergy Drug Targets. 2007;6(3):168173. doi:10.2174/187152807781696464

    • Search Google Scholar
    • Export Citation
  • 62.

    Marycz K, Szłapka-Kosarzewska J, Geburek F, Kornicka-Garbowska K. Systemic administration of rejuvenated adipose-derived mesenchymal stem cells improves liver metabolism in equine metabolic syndrome (EMS)–new approach in veterinary regenerative medicine. Stem Cell Rev Rep. 2019;15(6):842850. doi:10.1007/s12015-019-09913-3

    • Search Google Scholar
    • Export Citation
  • 63.

    Bruynsteen L, Erkens T, Peelman LJ, et al. Expression of inflammation-related genes is associated with adipose tissue location in horses. BMC Vet Res. 2013;9(1):240. doi:10.1186/1746-6148-9-240

    • Search Google Scholar
    • Export Citation
  • 64.

    Burns TA, Geor RJ, Mudge MC, McCutcheon LJ, Hinchcliff KW, Belknap JK. Proinflammatory cytokine and chemokine gene expression profiles in subcutaneous and visceral adipose tissue depots of insulin-resistant and insulin-sensitive light breed horses. J Vet Intern Med. 2010;24(4):932939. doi:10.1111/j.1939-1676.2010.0551.x

    • Search Google Scholar
    • Export Citation
  • 65.

    Waller AP, Huettner L, Kohler K, Lacombe VA. Novel link between inflammation and impaired glucose transport during equine insulin resistance. Vet Immunol Immunopathol. 2012;149(3–4):208215. doi:10.1016/j.vetimm.2012.07.003

    • Search Google Scholar
    • Export Citation
  • 66.

    Suagee JK, Corl BA, Crisman MV, Pleasant RS, Thatcher CD, Geor RJ. Relationships between body condition score and plasma inflammatory cytokines, insulin, and lipids in a mixed population of light-breed horses. J Vet Intern Med. 2013;27(1):157163. doi:10.1111/jvim.12021

    • Search Google Scholar
    • Export Citation
  • 67.

    Holbrook TC, Tipton T, McFarlane D. Neutrophil and cytokine dysregulation in hyperinsulinemic obese horses. Vet Immunol Immunopathol. 2012;145(1–2):283289. doi:10.1016/j.vetimm.2011.11.013

    • Search Google Scholar
    • Export Citation
  • 68.

    Zak A, Siwinska N, Elzinga S, et al. Effects of equine metabolic syndrome on inflammation and acute-phase markers in horses. Domest Anim Endocrinol. 2020;72:106448. doi:10.1016/j.domaniend.2020.106448

    • Search Google Scholar
    • Export Citation
  • 69.

    Treiber K, Carter R, Gay L, Williams C, Geor R. Inflammatory and redox status of ponies with a history of pasture-associated laminitis. Vet Immunol Immunopathol. 2009;129(3–4):216220. doi:10.1016/j.vetimm.2008.11.004

    • Search Google Scholar
    • Export Citation
  • 70.

    Vick MM, Adams AA, Murphy BA, et al. Relationships among inflammatory cytokines, obesity, and insulin sensitivity in the horse. J Anim Sci. 2007;85(5):11441155. doi:10.2527/jas.2006-673

    • Search Google Scholar
    • Export Citation
  • 71.

    Elzinga SE, Rohleder B, Schanbacher B, McQuerry K, Barker VD, Adams AA. Metabolic and inflammatory responses to the common sweetener stevioside and a glycemic challenge in horses with equine metabolic syndrome. Domest Anim Endocrinol. 2017;60:18. doi:10.1016/j.domaniend.2017.01.001

    • Search Google Scholar
    • Export Citation
  • 72.

    Dong J, Dong Y, Chen F, Mitch WE, Zhang L. Inhibition of myostatin in mice improves insulin sensitivity via irisin-mediated cross talk between muscle and adipose tissues. Int J Obes (Lond). 2016;40(3):434442. doi:10.1038/ijo.2015.200

    • Search Google Scholar
    • Export Citation
  • 73.

    Cleasby ME, Jarmin S, Eilers W, et al. Local overexpression of the myostatin propeptide increases glucose transporter expression and enhances skeletal muscle glucose disposal. Am J Physiol Endocrinol Metab. 2014;306(7):E814E823. doi:10.1152/ajpendo.00586.2013

    • Search Google Scholar
    • Export Citation
  • 74.

    Hill EW, Gu J, Eivers SS, et al. A sequence polymorphism in MSTN predicts sprinting ability and racing stamina in thoroughbred horses. PLoS One. 2010;5(1):e8645. doi:10.1371/journal.pone.0008645

    • Search Google Scholar
    • Export Citation
  • 75.

    Baron EE, Lopes MS, Mendonca D, da Camara Machado A. SNP identification and polymorphism analysis in exon 2 of the horse myostatin gene. Anim Genet. 2012;43(2):229232. doi:10.1111/j.1365-2052.2011.02229.x

    • Search Google Scholar
    • Export Citation
  • 76.

    Petersen JL, Valberg SJ, Mickelson JR, McCue ME. Haplotype diversity in the equine myostatin gene with focus on variants associated with race distance propensity and muscle fiber type proportions. Anim Genet. 2014;5(6):827835. doi:10.1111/age.12205

    • Search Google Scholar
    • Export Citation
  • 77.

    Takahashi H, Sato K, Yamaguchi T, et al. Myostatin alters glucose transporter-4 (GLUT4) expression in bovine skeletal muscles and myoblasts isolated from double-muscled (DM) and normal-muscled (NM) Japanese shorthorn cattle. Domest Anim Endocrinol. 2014;48:6268. doi:10.1016/j.domaniend.2014.01.007

    • Search Google Scholar
    • Export Citation
  • 78.

    Frank N, Tadros EM. Insulin dysregulation. Equine Vet J. 2014;46(1):103112. doi:10.1111/evj.12169

  • 79.

    Schuver A. Frank N, Chameroy KA, Elliot SB. Assessment of insulin and glucose dynamics by using an oral sugar test in horses. J Equine Vet Sci. 2014;34(4):465470. doi:10.1016/j.jevs.2013.09.006

    • Search Google Scholar
    • Export Citation
  • 80.

    Geor RJ. Current concepts on the pathophysiology of pasture-associated laminitis. Vet Clin North Am Equine Pract. 2010;26(2):26576. doi:10.1016/j.cveq.2010.06.001

    • Search Google Scholar
    • Export Citation
  • 81.

    Carter RA, McCutcheon LJ, Valle E, Meilahn EN, Geor RJ. Effects of exercise training on adiposity, insulin sensitivity, and plasma hormone and lipid concentrations in overweight or obese, insulin-resistant horses. Am J Vet Res. 2010;71(3):314321. doi:10.2460/ajvr.71.3.314

    • Search Google Scholar
    • Export Citation
  • 82.

    Bamford NJ, Potter SJ, Baskerville CL, Harris PA, Bailey SR. Effect of increased adiposity on insulin sensitivity and adipokine concentrations in different equine breeds adapted to cereal-rich or fat-rich meals. Vet J. 2016;214:1420. doi:10.1016/j.tvjl.2016.02.002

    • Search Google Scholar
    • Export Citation
  • 83.

    Yadav A, Kataria MA, Saini V. Role of leptin and adiponectin in insulin resistance. Clin Chim Acta. 2013;417:8084. doi:10.1016/j.cca.2012.12.007

    • Search Google Scholar
    • Export Citation
  • 84.

    Deng Y, Scherer PE. Adipokines as novel biomarkers and regulators of the metabolic syndrome. Ann N Y Acad Sci. 2010;1212:E1E19. doi:10.1111/j.1749-6632.2010.05875.x

    • Search Google Scholar
    • Export Citation
  • 85.

    Menzies-Gow NJ, Harris PA, Elliott J. Prospective cohort study evaluating risk factors for the development of pasture-associated laminitis in the United Kingdom. Equine Vet J. 2016;49(3):300306. doi:10.1111/evj.12606

    • Search Google Scholar
    • Export Citation
  • 86.

    Rivero JL, van Breda E, Rogers CW, Lindner A, van Oldruitenborgh-Oosterbaan MM. Unexplained underperformance syndrome in sport horses: classification, potential causes and recognition. Equine Vet J. 2008;40(6):611618. doi:10.2746/042516408x299118

    • Search Google Scholar
    • Export Citation
  • 87.

    Cadegiani FA, Kater CE. Hypothalamic-pituitary-adrenal (HPA) axis functioning in overtraining syndrome: findings from endocrine and metabolic responses on overtraining syndrome (EROS)-EROS-HPA axis. Sports Med Open. 2017;3(1):45. doi:10.1186/s40798-017-0113-0

    • Search Google Scholar
    • Export Citation
  • 88.

    de Graaf-Roelfsema E, Keizer HA, van Breda E, Wijnberg ID, van der Kolk JH. Hormonal responses to acute exercise, training and overtraining. A review with emphasis on the horse. Vet Q. 2007;29(3):82101. doi:10.1080/01652176.2007.9695232

    • Search Google Scholar
    • Export Citation
  • 89.

    Cheng AJ, Jude B, Lanner JT. Intramuscular mechanisms of overtraining. Redox Biol. 2020;35:101480. doi:10.1016/j.redox.2020.101480

  • 90.

    Cadegiani FA, Kater CE. Hormonal aspects of overtraining syndrome: a systematic review. BMC Sports Sci Med Rehabil. 2017;9:14. doi:10.1186/s13102-017-0079-8

    • Search Google Scholar
    • Export Citation
  • 91.

    Collins KH, Herzog W, MacDonald GZ, et al. Obesity, metabolic syndrome, and musculoskeletal disease: common inflammatory pathways suggest a central role for loss of muscle integrity. Front Physiol. 2018;9:112. doi:10.3389/fphys.2018.00112

    • Search Google Scholar
    • Export Citation
  • 92.

    Banse HE, Holbrook TC, Frank N, McFarlane D. Relationship of skeletal muscle inflammation with obesity and obesity-associated hyperinsulinemia in horses. Can J Vet Res. 2016;80(3):217224.

    • Search Google Scholar
    • Export Citation
  • 93.

    Stubbs NC, Kaiser LJ, Hauptman J, Clayton HM. Dynamic mobilisation exercises increase cross sectional area of musculus multifidus. Equine Vet J. 2011;43(5):522529. doi:10.1111/j.2042-3306.2010.00322.x

    • Search Google Scholar
    • Export Citation
  • 94.

    Nagy A, Murray JK, Dyson S. Elimination from elite endurance rides in nine countries: a preliminary study. Equine Vet J Suppl. 2010;(38):637643. doi:10.1111/j.2042-3306.2010.00220.x

    • Search Google Scholar
    • Export Citation
  • 95.

    Jansson A, Gunnarsson V, Ringmark S, et al. Increased body fat content in horses alters metabolic and physiological exercise response, decreases performance, and increases locomotion asymmetry. Physiol Rep. 2021;9(11):e14824. doi:10.14814/phy2.14824

    • Search Google Scholar
    • Export Citation
  • 96.

    Horsak B, Schwab C, Baca A, et al. Effects of a lower extremity exercise program on gait biomechanics and clinical outcomes in children and adolescents with obesity: a randomized controlled trial. Gait Posture. 2019;70:122129. doi:10.1016/j.gaitpost.2019.02.032

    • Search Google Scholar
    • Export Citation
  • 97.

    Shultz SP, D’Hondt E, Fink PW, Lenoir M, Hills AP. The effects of pediatric obesity on dynamic joint malalignment during gait. Clin Biomech (Bristol, Avon). 2014;29(7):835838. doi:10.1016/j.clinbiomech.2014.05.004

    • Search Google Scholar
    • Export Citation
  • 98.

    Li JS, Tsai TY, Clancy MM, Li G, Lewis CL, Felson DT. Weight loss changed gait kinematics in individuals with obesity and knee pain. Gait Posture. 2019;68:461465. doi:10.1016/j.gaitpost.2018.12.031

    • Search Google Scholar
    • Export Citation
  • 99.

    de Oliveira Máximo R, de Oliveira DC, Ramírez PC, et al. Dynapenia, abdominal obesity or both: which accelerates the gait speed decline most? Age Ageing. 2021;50(5):16161625. doi:10.1093/ageing/afab093

    • Search Google Scholar
    • Export Citation
  • 100.

    Sanchez-Santos MT, Judge A, Gulati M, et al. Association of metabolic syndrome with knee and hand osteoarthritis: a community-based study of women. Semin Arthritis Rheum. 2019;48(5):791798. doi:10.1016/j.semarthrit.2018.07.007

    • Search Google Scholar
    • Export Citation
  • 101.

    Valdes AM. Metabolic syndrome and osteoarthritis pain: common molecular mechanisms and potential therapeutic implications. Osteoarthritis Cartilage. 2020;28(1):79. doi:10.1016/j.joca.2019.06.015

    • Search Google Scholar
    • Export Citation
  • 102.

    Abate M, Schiavone C, Salini V, Andia I. Occurrence of tendon pathologies in metabolic disorders. Rheumatology (Oxford). 2013;52(4):599608. doi:10.1093/rheumatology/kes395

    • Search Google Scholar
    • Export Citation
  • 103.

    Oliva F, Marsilio E, Asparago G, et al. Achilles tendon rupture and dysmetabolic diseases: a multicentric, epidemiologic study. J Clin Med. 2022;11(13):3698. doi:10.3390/jcm11133698

    • Search Google Scholar
    • Export Citation
  • 104.

    Bay-Jensen AC, Slagboom E, Chen-An P, et al. Role of hormones in cartilage and joint metabolism: understanding an unhealthy metabolic phenotype in osteoarthritis. Menopause. 2012;20(5):578586. doi:10.1097/gme.0b013e3182745993

    • Search Google Scholar
    • Export Citation
  • 105.

    Jiang H, Pu Y, Li ZH, et al. Adiponectin, may be a potential protective factor for obesity-related osteoarthritis. Diabetes Metab Syndr Obes. 2022;15:13051319. doi:10.2147/dmso.s359330

    • Search Google Scholar
    • Export Citation
  • 106.

    Liu B, Gao YH, Dong N, et al. Differential expression of adipokines in the synovium and infrapatellar fat pad of osteoarthritis patients with and without metabolic syndrome. Connect Tissue Res. 2019;60(6):611618. doi:10.1080/03008207.2019.1620221

    • Search Google Scholar
    • Export Citation
  • 107.

    Schett G, Kleyer A, Perricone C, et al. Diabetes is an independent predictor for severe osteoarthritis: results from a longitudinal cohort study. Diabetes Care. 2013;36(2):403409. doi:10.2337/dc12-0924

    • Search Google Scholar
    • Export Citation
  • 108.

    Xie C, Chen Q. Adipokines: new therapeutic target for osteoarthritis? Curr Rheumatol Rep. 7 2019;21(12):71. doi:10.1007/s11926-019-0868-z

    • Search Google Scholar
    • Export Citation
  • 109.

    Boyce M, Trumble TN, Groschen DM, Merritt KA, Brown MP. Collagen biomarker response to acute joint injury in a non-terminal model of osteoarthritis. 2011.

  • 110.

    Boyce M, Trumble TN, Carlson CS, Groschen DM, Merritt KA, Brown MP. Non-terminal model of acute joint injury causes early osteoarthritis. Osteoarthritis and Cartilage. 2013;21(5):746755. doi:10.1016/j.oca.2013.02.653

    • Search Google Scholar
    • Export Citation
  • 111.

    Trumble TN, Groschen DM, Ha N, Boyce M, Merritt KA, Brown MP. New urine biomarker assay compares to synovial fluid changes after acute joint injury in an equine model of osteoarthritis. Osteoarthritis Cartilage. 2012;20(suppl 1):S91.

    • Search Google Scholar
    • Export Citation
  • 112.

    Le TK, Montejano LB, Cao Z, Zhao Y, Ang D. Health care costs in US patients with and without a diagnosis of osteoarthritis. J Pain Res. 2012;5:2330. doi:10.2147/jpr.s27275

    • Search Google Scholar
    • Export Citation
  • 113.

    Ray A, Ray BK. An inflammation-responsive transcription factor in the pathophysiology of osteoarthritis. Biorheology. 2008;45(3–4):399409. doi:10.3233/BIR-2008-0500

    • Search Google Scholar
    • Export Citation
  • 114.

    Watts AE, Dabareiner R, Marsh C, Carter GK, Cummings KJ. A randomized, controlled trial of the effects of resveratrol administration in performance horses with lameness localized to the distal tarsal joints. J Am Vet Med Assoc. 2016;249(6):650659. doi:10.2460/javma.249.6.650

    • Search Google Scholar
    • Export Citation
  • 115.

    Zhou Q, Wang Y, Han X, Fu S, Zhu C, Chen Q. Efficacy of resveratrol supplementation on glucose and lipid metabolism: a meta-analysis and systematic review. Front Physiol. 2022;13:795980. doi:10.3389/fphys.2022.795980

    • Search Google Scholar
    • Export Citation
  • 116.

    Chaplin A, Carpéné C, Mercader J. Resveratrol, metabolic syndrome, and gut microbiota. Nutrients. 2018;10(11):1651. doi:10.3390/nu10111651

    • Search Google Scholar
    • Export Citation
  • 117.

    Wang P, Li D, Ke W, Liang D, Hu X, Chen F. Resveratrol-induced gut microbiota reduces obesity in high-fat diet-fed mice. Int J Obes (Lond). 2019;44(1):213225. doi:10.1038/s41366-019-0332-1

    • Search Google Scholar
    • Export Citation
  • 118.

    Kosolofski HR, Gow SP, Robinson KA. Prevalence of obesity in the equine population of Saskatoon and surrounding area. Can Vet J. 2017;58(9):967970.

    • Search Google Scholar
    • Export Citation
  • 119.

    Giles SL, Rands SA, Nicol CJ, Harris PA. Obesity prevalence and associated risk factors in outdoor living domestic horses and ponies. PeerJ. 2014;2:e299. doi:10.7717/peerj.299

    • Search Google Scholar
    • Export Citation
  • 120.

    Gluck C, Williams J, Pratt-Phillips S. Performance analysis of show jumping rounds at a national pony competition. Comp Exerc Physiol; 2022;18(1):S13.

    • Search Google Scholar
    • Export Citation
  • 121.

    Tang Q, Hu ZC, Shen LY, Shang P, Xu HZ, Liu HX. Association of osteoarthritis and circulating adiponectin levels: a systematic review and meta-analysis. Lipids Health Dis. 2018;17(1):189. doi:10.1186/s12944-018-0838-x

    • Search Google Scholar
    • Export Citation
  • 122.

    Zhang P, Zhong ZH, Yu HT, Liu B. Significance of increased leptin expression in osteoarthritis patients. PLoS One. 2015;10(4):e0123224. doi:10.1371/journal.pone.0123224

    • Search Google Scholar
    • Export Citation
  • 123.

    Dong N, Gao YH, Liu B, et al. Differential expression of adipokines in knee osteoarthritis patients with and without metabolic syndrome. Int Orthop. 2018;42(6):12831289. doi:10.1007/s00264-018-3761-x

    • Search Google Scholar
    • Export Citation
  • 124.

    Fuentes-Romero B, Muñoz-Prieto A, Cerón JJ, et al. Measurement of plasma resistin concentrations in horses with metabolic and inflammatory disorders. Animals (Basel). 2021;12(1):77. doi:10.3390/ani12010077

    • Search Google Scholar
    • Export Citation
  • 125.

    Brown MP, Trumble TN, Merritt KA. High-mobility group box chromosomal protein 1 as a potential inflammatory biomarker of joint injury in Thoroughbreds. Am J Vet Res. 2009;70(10):12301235. doi:10.2460/ajvr.70.10.1230

    • Search Google Scholar
    • Export Citation
  • 126.

    Trumble TN, Scarbrough AB, Brown MP. Osteochondral injury increases type II collagen degradation products (C2C) in synovial fluid of Thoroughbred racehorses. Osteoarthritis Cartilage. 2009;17(3):371374. doi:10.1016/j.joca.2008.07.014

    • Search Google Scholar
    • Export Citation
  • 127.

    McIlwraith CW, Kawcak CE, Frisbie DD, et al. Biomarkers for equine joint injury and osteoarthritis. J Orthop Res. 2018;36(3):823831. doi:10.1002/jor.23738

    • Search Google Scholar
    • Export Citation
  • 128.

    Skovgaard D, Siersma VD, Klausen SB, et al. Chronic hyperglycemia, hypercholesterolemia, and metabolic syndrome are associated with risk of tendon injury. Scand J Med Sci Sports. 2021;31(9):18221831. doi:10.1111/sms.13984

    • Search Google Scholar
    • Export Citation
  • 129.

    Hofberger SC, Gauff F, Thaller D, Morgan R, Keen JA, Licka TF. Assessment of tissue-specific cortisol activity with regard to degeneration of the suspensory ligaments in horses with pituitary pars intermedia dysfunction. Am J Vet Res. 2018;79(2):199210. doi:10.2460/ajvr.79.2.199

    • Search Google Scholar
    • Export Citation
  • 130.

    Mousa A, Jones S, Toft A, Perros P. Spontaneous rupture of Achilles tendon: missed presentation of Cushing’s syndrome. BMJ. 28 1999;319(7209):560561. doi:10.1136/bmj.319.7209.560

    • Search Google Scholar
    • Export Citation
  • 131.

    Batisse M, Somda F, Delorme JP, Desbiez F, Thieblot P, Tauveron I. Spontaneous rupture of Achilles tendon and Cushing’s disease. Case report. Ann Endocrinol (Paris). 2008;69(6):530531. doi:10.1016/j.ando.2008.06.003

    • Search Google Scholar
    • Export Citation
  • 132.

    Yamaoka K, Tango T. Effects of lifestyle modification on metabolic syndrome: a systematic review and meta-analysis. BMC Med. 2012;10:138. doi:10.1186/1741-7015-10-138

    • Search Google Scholar
    • Export Citation
  • 133.

    Riddle DL, Stratford PW. Body weight changes and corresponding changes in pain and function in persons with symptomatic knee osteoarthritis: a cohort study. Arthritis Care Res (Hoboken). 2013;65(1):1522. doi:10.1002/acr.21692

    • Search Google Scholar
    • Export Citation
  • 134.

    Delarocque J, Frers F, Huber K, Feige K, Warnken T. Weight loss is linearly associated with a reduction of the insulin response to an oral glucose test in Icelandic horses. BMC Vet Res. 2020;16(1):151. doi:10.1186/s12917-020-02356-w

    • Search Google Scholar
    • Export Citation
  • 135.

    Bamford NJ, Potter SJ, Baskerville CL, Harris PA, Bailey SR. Influence of dietary restriction and low-intensity exercise on weight loss and insulin sensitivity in obese equids. J Vet Intern Med. 2019;33(1):280286. doi:10.1111/jvim.15374

    • Search Google Scholar
    • Export Citation
  • 136.

    Freestone JF, Beadle R, Shoemaker K, Bessin RT, Wolfsheimer KJ, Church C. Improved insulin sensitivity in hyperinsulinaemic ponies through physical conditioning and controlled feed intake. Equine Vet J. 1992;24(3):187190. doi:10.1111/j.2042-3306.1992.tb02812.x

    • Search Google Scholar
    • Export Citation
  • 137.

    Normandin E, Chmelo E, Lyles MF, Marsh AP, Nicklas BJ. Effect of resistance training and caloric restriction on the metabolic syndrome. Med Sci Sports Exerc. 2017;49(3):413419. doi:10.1249/mss.0000000000001122

    • Search Google Scholar
    • Export Citation
  • 138.

    Bakker EA, Lee DC, Sui X, et al. Association of resistance exercise, independent of and combined with aerobic exercise, with the incidence of metabolic syndrome. Mayo Clin Proc. 2017;92(8):12141222. doi:10.1016/j.mayocp.2017.02.018

    • Search Google Scholar
    • Export Citation
  • 139.

    Pfau T, Simons V, Rombach N, Stubbs N, Weller R. Effect of a 4-week elastic resistance band training regimen on back kinematics in horses trotting in-hand and on the lunge. Equine Vet J. 2017;49(6):829835. doi:10.1111/evj.12690

    • Search Google Scholar
    • Export Citation
  • 140.

    Ursini T, Shaw K, Levine D, Richards J, Adair HS. Electromyography of the multifidus muscle in horses trotting during therapeutic exercises. Front Vet Sci. 2022;9:844776. doi:10.3389/fvets.2022.844776

    • Search Google Scholar
    • Export Citation
  • 141.

    Miller MA, Croft LB, Belanger AR, et al. Prevalence of metabolic syndrome in retired National Football League players. Am J Cardiol. 2008;101(9):12811284. doi:10.1016/j.amjcard.2007.12.029

    • Search Google Scholar
    • Export Citation
  • 142.

    Nostell K, Lindåse S, Edberg H, Bröjer J. The effect of insulin infusion on heart rate and systemic blood pressure in horses with equine metabolic syndrome. Equine Vet J. 2019;51(6):733737. doi:10.1111/evj.13110

    • Search Google Scholar
    • Export Citation
  • 143.

    Ali AT. Polycystic ovary syndrome and metabolic syndrome. Ceska Gynekol. 2015;80(4):279289.

  • 144.

    Broughton DE, Moley KH. Obesity and female infertility: potential mediators of obesity’s impact. Fertil Steril. 2017;107(4):840847. doi:10.1016/j.fertnstert.2017.01.017

    • Search Google Scholar
    • Export Citation
  • 145.

    Sessions-Bresnahan DR, Schauer KL, Heuberger AL, Carnevale EM. Effect of obesity on the preovulatory follicle and lipid fingerprint of equine oocytes. Biol Reprod. 2016;94(1):15. doi:10.1095/biolreprod.115.130187

    • Search Google Scholar
    • Export Citation
  • 146.

    Sessions-Bresnahan DR, Heuberger AL, Carnevale EM. Obesity in mares promotes uterine inflammation and alters embryo lipid fingerprints and homeostasis. Biol Reprod. 2018;99(4):761772. doi:10.1093/biolre/ioy107

    • Search Google Scholar
    • Export Citation
  • 147.

    Burns TA. Effects of common equine endocrine diseases on reproduction. Vet Clin North Am Equine Pract. 2016;32(3):435449. doi:10.1016/j.cveq.2016.07.005

    • Search Google Scholar
    • Export Citation
  • 148.

    Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiol Rev. 2014;94(4):10271076. doi:10.1152/physrev.00029.2013

    • Search Google Scholar
    • Export Citation
  • 149.

    Robles M, Hammer C, Staniar B, Chavatte-Palmer P. Nutrition of broodmares. Vet Clin North Am Equine Pract. 2021;37(1):177205. doi:10.1016/j.cveq.2021.01.001

    • Search Google Scholar
    • Export Citation
  • 150.

    Peugnet P, Robles M, Wimel L, Tarrade A, Chavatte-Palmer P. Management of the pregnant mare and long-term consequences on the offspring. Theriogenology. 2016;86(1):99109. doi:10.1016/j.theriogenology.2016.01.028

    • Search Google Scholar
    • Export Citation
  • 151.

    Robles M, Gautier C, Mendoza L, et al. Maternal nutrition during pregnancy affects testicular and bone development, glucose metabolism and response to overnutrition in weaned horses up to two years. PLoS One. 2017;12(1):e0169295. doi:10.1371/journal.pone.0169295

    • Search Google Scholar
    • Export Citation
  • 152.

    Thorson JF, Karren BJ, Bauer ML, Cavinder CA, Coverdale JA, Hammer CJ. Effect of selenium supplementation and plane of nutrition on mares and their foals: foaling data. J Anim Sci. 2010;88(3):982990. doi:10.2527/jas.2008-1646

    • Search Google Scholar
    • Export Citation
  • 153.

    Gorgal R, Gonçalves E, Barros M, et al. Gestational diabetes mellitus: a risk factor for non-elective cesarean section. J Obstet Gynaecol Res. 2012;38(1):154159. doi:10.1111/j.1447-0756.2011.01659.x

    • Search Google Scholar
    • Export Citation
  • 154.

    Daskalakis G, Marinopoulos S, Krielesi V, et al. Placental pathology in women with gestational diabetes. Acta Obstet Gynecol Scand. 2008;87(4):403407. doi:10.1080/00016340801908783

    • Search Google Scholar
    • Export Citation
  • 155.

    Coverdale JA, Hammer CJ, Walter KW. Horse species symposium: nutritional programming and the impact on mare and foal performance. J Anim Sci. 2015;93(7):3261 73267. doi:10.2527/jas.2015-9057

    • Search Google Scholar
    • Export Citation
  • 156.

    Gluckman PD, Hanson MA. Developmental and epigenetic pathways to obesity: an evolutionary-developmental perspective. Int J Obes (Lond). 2008;32(suppl 7):S62S71. doi:10.1038/ijo.2008.240

    • Search Google Scholar
    • Export Citation
  • 157.

    Norton EM, Schultz NE, Rendahl AK, et al. Heritability of metabolic traits associated with equine metabolic syndrome in Welsh ponies and Morgan horses. Equine Vet J. 2019;51(4):475480. doi:10.1111/evj.13053

    • Search Google Scholar
    • Export Citation
  • 158.

    Lewis SL, Holl HM, Streeter C, et al. Genomewide association study reveals a risk locus for equine metabolic syndrome in the Arabian horse. J Anim Sci. 2017;95(3):10711079. doi:10.2527/jas.2016.1221

    • Search Google Scholar
    • Export Citation
  • 159.

    Norton EM, Avila F, Schultz NE, Mickelson JR, Geor RJ, McCue ME. Evaluation of an HMGA2 variant for pleiotropic effects on height and metabolic traits in ponies. J Vet Intern Med. 2019;33(2):942952. doi:10.1111/jvim.15403

    • Search Google Scholar
    • Export Citation
  • 160.

    Cash CM, Fitzgerald DM, Spence RJ, de Laat MA. Preliminary analysis of the FAM174A gene suggests it lacks a strong association with equine metabolic syndrome in ponies. Domest Anim Endocrinol. 2020;72:106439. doi:10.1016/j.domaniend.2020.106439

    • Search Google Scholar
    • Export Citation
  • 161.

    Roy MM, Norton EM, Rendahl AK, et al. Assessment of the FAM174A 11G allele as a risk allele for equine metabolic syndrome. Anim Genet. 2020;51(4):607610. doi:10.1111/age.12952

    • Search Google Scholar
    • Export Citation
  • 162.

    Norton E, Schultz N, Geor R, McFarlane D, Mickelson J, McCue M. Genome-wide association analyses of equine metabolic syndrome phenotypes in welsh ponies and morgan horses. Genes (Basel). 2019;10(11):893. doi:10.3390/genes10110893

    • Search Google Scholar
    • Export Citation
  • 163.

    Schaefer RJ, Cullen J, Manfredi J, McCue M. Functional contexts of adipose and gluteal muscle tissue gene co-expression networks in the domestic horse. Integr Comp Biol. 2020:icaa134. doi:10.1093/icb/icaa134

    • Search Google Scholar
    • Export Citation
  • 164.

    Delarocque J, Reiche DB, Meier AD, Warnken T, Feige K, Sillence MN. Metabolic profile distinguishes laminitis-susceptible and -resistant ponies before and after feeding a high sugar diet. BMC Vet Res. 2021;17(1):56. doi:10.1186/s12917-021-02763-7

    • Search Google Scholar
    • Export Citation
  • 165.

    Jacob SI, Murray KJ, Rendahl AK, Geor RJ, Schultz NE, McCue ME. Metabolic perturbations in Welsh Ponies with insulin dysregulation, obesity, and laminitis. J Vet Intern Med. 2018;32(3):12151233. doi:10.1111/jvim.15095

    • Search Google Scholar
    • Export Citation
  • 166.

    Patterson Rosa L, Mallicote MF, Long MT, Brooks SA. Metabogenomics reveals four candidate regions involved in the pathophysiology of Equine Metabolic Syndrome. Mol Cell Probes. 2020;53:101620. doi:10.1016/j.mcp.2020.101620

    • Search Google Scholar
    • Export Citation
  • 167.

    Rexroad C, Vallet J, Matukumalli LK, et al. Genome to phenome: improving animal health, production, and well-being–a new USDA blueprint for animal genome research 2018–2027. Front Genet. 2019;10:327. doi:10.3389/fgene.2019.00327

    • Search Google Scholar
    • Export Citation
  • 168.

    Elliott HR, Sharp GC, Relton CL, Lawlor DA. Epigenetics and gestational diabetes: a review of epigenetic epidemiology studies and their use to explore epigenetic mediation and improve prediction. Diabetologia. 2019;62(12):21712178. doi:10.1007/s00125-019-05011-8

    • Search Google Scholar
    • Export Citation
  • 169.

    Allard C, Desgagné V, Patenaude J, et al. Mendelian randomization supports causality between maternal hyperglycemia and epigenetic regulation of leptin gene in newborns. Epigenetics. 2015;10(4):342351. doi:10.1080/15592294.2015.1029700

    • Search Google Scholar
    • Export Citation
  • 170.

    Zhang A, Sun H, Wang X. Serum metabolomics as a novel diagnostic approach for disease: a systematic review. Anal Bioanal Chem. 2012;404(4):12391245. doi:10.1007/s00216-012-6117-1

    • Search Google Scholar
    • Export Citation
  • 171.

    Batch BC, Shah SH, Newgard CB, et al. Branched chain amino acids are novel biomarkers for discrimination of metabolic wellness. Metabolism. 2013;62(7):961969. doi:10.1016/j.metabol.2013.01.007

    • Search Google Scholar
    • Export Citation
  • 172.

    Menni C, Fauman E, Erte I, et al. Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes. 2013;62(12):42704276. doi:10.2337/db13-0570

    • Search Google Scholar
    • Export Citation
  • 173.

    Walford GA, Davis J, Warner AS, et al. Branched chain and aromatic amino acids change acutely following two medical therapies for type 2 diabetes mellitus. Metabolism. 2013;62(12):17721778. doi:10.1016/j.metabol.2013.07.003

    • Search Google Scholar
    • Export Citation
  • 174.

    Huffman KM, Shah SH, Stevens RD, et al. Relationships between circulating metabolic intermediates and insulin action in overweight to obese, inactive men and women. Diabetes Care. 2009;32(9):16781683. doi:10.2337/dc08-2075

    • Search Google Scholar
    • Export Citation
  • 175.

    Chen Q, Wang W, Xia MF, et al. Identification of circulating sphingosine kinase-related metabolites for prediction of type 2 diabetes. J Transl Med. 2021;19(1):393. doi:10.1186/s12967-021-03066-z

    • Search Google Scholar
    • Export Citation
  • 176.

    Floegel A, Stefan N, Yu Z, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62(2):639648. doi:10.2337/db12-0495

    • Search Google Scholar
    • Export Citation
  • 177.

    Milburn MV, Lawton KA. Application of metabolomics to diagnosis of insulin resistance. Annu Rev Med. 2013;64:291305. doi:10.1146/annurev-med-061511-134747

    • Search Google Scholar
    • Export Citation
  • 178.

    Gall WE, Beebe K, Lawton KA, et al. Alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One. 2010;5(5):e10883. doi:10.1371/journal.pone.0010883

    • Search Google Scholar
    • Export Citation
  • 179.

    Ho JE, Larson MG, Vasan RS, et al. Metabolite profiles during oral glucose challenge. Diabetes. 2013;62(8):26892698. doi:10.2337/db12-0754

    • Search Google Scholar
    • Export Citation
  • 180.

    Bain JR, Muehlbauer MJ. Metabolomics reveals unexpected responses to oral glucose. Diabetes. 2013;62(8):26512653. doi:10.2337/db13-0605

    • Search Google Scholar
    • Export Citation
  • 181.

    Coleman MC, Whitfield-Cargile CM, Madrigal RG, Cohen ND. Comparison of the microbiome, metabolome, and lipidome of obese and non-obese horses. PLoS One. 2019;14(4):e0215918. doi:10.1371/journal.pone.0215918

    • Search Google Scholar
    • Export Citation
  • 182.

    Klein DJ, Anthony TG, McKeever KH. Metabolomics in equine sport and exercise. J Anim Physiol Anim Nutr (Berl). 2021;105(1):140148. doi:10.1111/jpn.13384

    • Search Google Scholar
    • Export Citation
  • 183.

    Klein DJ, McKeever KH, Mirek ET, Anthony TG. Metabolomic response of equine skeletal muscle to acute fatiguing exercise and training. Front Physiol. 2020;11:110. doi:10.3389/fphys.2020.00110

    • Search Google Scholar
    • Export Citation
  • 184.

    Savikj M, Stocks B, Sato S, et al. Exercise timing influences multi-tissue metabolome and skeletal muscle proteome profiles in type 2 diabetic patients–a randomized crossover trial. Metabolism. 2022;135:155268. doi:10.1016/j.metabol.2022.155268

    • Search Google Scholar
    • Export Citation
  • 185.

    Mancilla R, Brouwers B, Schrauwen-Hinderling VB, Hesselink MKC, Hoeks J, Schrauwen P. Exercise training elicits superior metabolic effects when performed in the afternoon compared to morning in metabolically compromised humans. Physiol Rep. 2021;8(24):e14669. doi:10.14814/phy2.14669

    • Search Google Scholar
    • Export Citation
  • 186.

    Stefaniuk M, Ropka-Molik K. RNA sequencing as a powerful tool in searching for genes influencing health and performance traits of horses. J Appl Genet. 2016;57(2):199206. doi:10.1007/s13353-015-0320-7

    • Search Google Scholar
    • Export Citation
  • 187.

    Manfredi JM. Identifying Breed Differences in Insulin Dynamics, Skeletal Muscle and Adipose Tissue Histology, and Gene Expression. Michigan State University; 2016.

    • Search Google Scholar
    • Export Citation
  • 188.

    Ropka-Molik K, Stefaniuk-Szmukier M, Żukowski K, Piórkowska K, Gurgul A, Bugno-Poniewierska M. Transcriptome profiling of Arabian horse blood during training regimens. BMC Genet. 2017;18(1):31. doi:10.1186/s12863-017-0499-1

    • Search Google Scholar
    • Export Citation
  • 189.

    Kogelman LJ, Fu J, Franke L, et al. Inter-tissue gene co-expression networks between metabolically healthy and unhealthy obese individuals. PLoS One. 2016;11(12):e0167519. doi:10.1371/journal.pone.0167519

    • Search Google Scholar
    • Export Citation
  • 190.

    Pennington PM, Splan RK, Jacobs RD, et al. Influence of metabolic status and diet on early pregnant equine histotroph proteome: preliminary findings. J Equine Vet Sci. 2020;88:102938. doi:10.1016/j.jevs.2020.102938

    • Search Google Scholar
    • Export Citation
  • 191.

    Henry ML, Velez-Irizarry D, Pagan JD, Sordillo L, Gandy J, Valberg SJ. The impact of N-acetyl cysteine and coenzyme Q10 supplementation on skeletal muscle antioxidants and proteome in fit thoroughbred horses. Antioxidants (Basel). 2021;10(11):1739. doi:10.3390/antiox10111739

    • Search Google Scholar
    • Export Citation
  • 192.

    Peffers MJ, Beynon RJ, Clegg PD. Absolute quantification of selected proteins in the human osteoarthritic secretome. Int J Mol Sci. 2013;14(10):2065820681. doi:10.3390/ijms141020658

    • Search Google Scholar
    • Export Citation
  • 193.

    Daugaard JR, Richter EA. Relationship between muscle fibre composition, glucose transporter protein 4 and exercise training: possible consequences in non-insulin-dependent diabetes mellitus. Acta Physiol Scand. 2001;171(3):267276. doi:10.1046/j.1365-201x.2001.00829.x

    • Search Google Scholar
    • Export Citation
  • 194.

    Saad MJ, Santos A, Prada PO. Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda). 2016;31(4):283293. doi:10.1152/physiol.00041.2015

    • Search Google Scholar
    • Export Citation
  • 195.

    Binvignat M, Sokol H, Mariotti-Ferrandiz E, Berenbaum F, Sellam J. Osteoarthritis and gut microbiome. Joint Bone Spine. 2021;88(5):105203. doi:10.1016/j.jbspin.2021.105203

    • Search Google Scholar
    • Export Citation
  • 196.

    Dougal K, Harris PA, Edwards A, et al. A comparison of the microbiome and the metabolome of different regions of the equine hindgut. FEMS Microbiol Ecol. 2012;82(3):642652. doi:10.1111/j.1574-6941.2012.01441.x

    • Search Google Scholar
    • Export Citation
  • 197.

    Dougal K, de la Fuente G, Harris PA, et al. Characterisation of the faecal bacterial community in adult and elderly horses fed a high fibre, high oil or high starch diet using 454 pyrosequencing. PLoS One. 2014;9(2):e87424. doi:10.1371/journal.pone.0087424

    • Search Google Scholar
    • Export Citation
  • 198.

    Costa MC, Weese JS. Understanding the intestinal microbiome in health and disease. Vet Clin North Am Equine Pract. 2018;34(1):112. doi:10.1016/j.cveq.2017.11.005

    • Search Google Scholar
    • Export Citation
  • 199.

    Fernandes KA, Kittelmann S, Rogers CW, et al. Faecal microbiota of forage-fed horses in New Zealand and the population dynamics of microbial communities following dietary change. PLoS One. 2014;9(11):e112846. doi:10.1371/journal.pone.0112846

    • Search Google Scholar
    • Export Citation
  • 200.

    Elzinga SE, Weese JS, Adams AA. Comparison of the fecal microbiota in horses with equine metabolic syndrome and metabolically normal controls fed a similar diet. J Equine Vet Sci. 2016;44:916.

    • Search Google Scholar
    • Export Citation
All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 6496 3725 416
PDF Downloads 2748 709 36
Advertisement