ReviewTranslational Physiology

High-fidelity porcine models of metabolic syndrome: a contemporary synthesis

Published Online:https://doi.org/10.1152/ajpendo.00413.2021

Abstract

This review aims to describe and compare porcine models of metabolic syndrome. This syndrome and its associated secondary comorbidities are set to become the greatest challenge to healthcare providers and policy makers in the coming century. However, an incomplete understanding of the pathogenesis has left significant knowledge gaps in terms of efficacious therapeutics. To further our comprehension and, in turn, management of metabolic syndrome, appropriate high-fidelity models of the disease complex are of great importance. In this context, our review aims to assess the most promising porcine models of metabolic syndrome currently available for their similarity to the human phenotype. In addition, we aim to highlight the strengths and shortcomings of each model in an attempt to identify the most appropriate application of each. Although no porcine model perfectly recapitulates the human metabolic syndrome, several pose satisfactory approximations. The Ossabaw miniature swine in particular represents a highly translatable model that develops each of the core parameters of the syndrome with many of the associated secondary comorbidities. Future high-fidelity porcine models of metabolic syndrome need to focus on secondary sequelae replication, which may require extended induction period to reveal.

INTRODUCTION

Metabolic syndrome (MetS) is an interrelated cluster of metabolic dysfunctions and risk factors which arises as a direct result of genetics, environmental exposures, and lifestyle choices. Defining criteria have varied between health organizations, but diagnosis commonly necessitates some degree of truncal obesity, in combination with type 2 diabetes, insulin resistance, dyslipidemia, or hypertension (Fig. 1) (1). We are only now beginning to identify and understand the downstream links with anatomic structural remodeling processes and important secondary comorbidities during MetS, such as hepatic steatosis, periorgan adiposity, heart dysfunction, and end-organ failure. Of the secondary functional changes, perhaps one of the most compelling for the coming decades is the inflammation and subsequent remodeling of cardiac tissue with impaired pump function, which in turn can lead to heart failure and arrhythmia (2).

Figure. 1.

Figure. 1.Metabolic syndrome core components and secondary end-organ sequelae in porcine models. HF-pEF, heart failure with preserved ejection fraction; MetS, metabolic syndrome; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; TAG, triacylglycerol. Created using Biorender.com.


Despite the propensity for expansion of MetS superpathology across global populations, a satisfactory understanding of this relatively complex cardiometabolic disease state is still lacking. Therefore, physicians and consequently, their patients have a dearth of effective therapies available to them which go beyond symptom management. However, given the current global metabolic syndrome trajectory, with exponentially rising obesity rates (3), an ever-expanding elderly population (4) and inadequate preventive therapies for heart failure with preserved ejection fraction (HF-pEF) and atrial fibrillation, these secondary pathologies are likely to quickly become the central scourge of Western healthcare systems for decades to come. To further our understanding and thus provide ensuing therapeutic options for the management of MetS and its associated comorbidities, it is essential that we develop high-fidelity animal models which not only display similar pathophenotype, but respond similarly to known disease state modifiers. Sus scrofa as a species offers an excellent degree of similarity to human physiology. In addition, pigs as omnivores are amenable to different dietary programs, including the “Western”-type diet—rich in carbohydrates, saturated fats, cholesterol and salt—which is central to the instigation of MetS (5). These two characteristics make the species an extremely useful model for metabolic disease research and therapeutic development.

This review aims to present a contemporary update of the studies which have used porcine models of MetS. In addition, we aim to critically appraise and contrast the relative strengths of each in an attempt to create a comprehensive reference material for metabolic researchers seeking to select the model most appropriate to their study design and objectives. Finally, we aim to highlight the advantages of porcine models of MetS and identify their current shortcomings and limitations to guide future development.

METHODS

Search Strategy and Criteria

MEDLINE, SCOPUS, and PUBMED databases were searched using the following keywords: “Swine,” “Pig,” “Porcine,” “Model,” “Metabolic Syndrome,” “Metabolic dysfunction.” To ensure the focus was maintained on the most current data, only studies published following 01 January 2005 were included in this review and the final update search was conducted in June 2021. Review articles were entirely excluded from the initial searches and only English language publications were considered. We intended to include every study in which a group of pigs (population) was treated with a diet, drug, or procedure (intervention) that recapitulated the features of MetS (outcome), when compared with untreated animals or baseline (control). The search strategy limited the results to articles which explicitly indicated that a pig model of MetS was applied or created. However, several models that met the definition of MetS were also included, although they were not explicitly deemed so within their respective reports and, as such, would otherwise not be uncovered by the search approach. Studies that lacked a comparative control or did not report on the core parameters of interest were excluded.

Data Extraction

All studies that were ultimately included in the final review were assessed for animal breed and inducing factors (diet, drugs or procedures), as well as study aims and outcomes. In addition, each study was assessed for presence or absence of primary and secondary features of MetS displayed by the model, including obesity, hypertension, insulin resistance, glucose intolerance, hypercholesterolemia, hypertriglyceridemia, microbiota alterations, intestinal permeability, fatty liver disease, systemic and adipose tissue inflammation, cardiac disease, and neurological disorder. Studies that exclusively focused on atherosclerosis and vascular disease were excluded since these aspects of MetS have been extensively reviewed elsewhere (68).

RESULTS

Search Returns

The initial search strategy uncovered 72 studies of relevance in MEDLINE, 243 studies of relevance in SCOPUS, 76 studies of relevance in PUBMED, with 25 additional studies added during unstructured reviewing of the literature and reference lists. After removal of duplicates, the titles and abstracts of 379 studies were screened for suitability, after which 144 articles remained. Finally, 17 more articles were excluded due to model unsuitability, leaving 127 articles describing the use of a porcine model of MetS. Seven of the included articles reported on Bama minipig (915), 1 on Chinese miniature pig (16), 21 on the domestic pig (1737), 2 on Duroc pig (38, 39), 1 on Duroc × Landrace × Yorkshire pig (40), 12 on Göttingen minipig (4152), 4 on Iberian pig (5356), 4 on Landrace pig (55, 5759), 5 on Lee-Sung and Lanyu minipigs (6064), 3 on Microminipig (6567), 46 on Ossabaw miniature swine (51, 68112), 12 on Yorkshire pig (18, 92, 113123), 4 on Yorkshire × Landrace cross pig (124127), 6 on Yucatan minipig (90, 128132), and 1 on Tibetan minipig (133). A summary of the metabolic disease profile of the identified models is presented in Fig. 2. It is of note that this figure does not include the vascular and atherosclerotic consequences of MetS to avoid repetition with other reviews (6). In addition, representative values of MetS core components for each porcine model are provided in Table 1.

Figure 2.

Figure 2.Summary of disease profile of known porcine models of metabolic syndrome. AB-HT, aortic-banding-induced hypertension; BF, body fat; BW, body weight; DOCA-HT, deoxycorticosteroneactetate-induced hypertension; GIT, glucose intolerance; HT, hypertension; IR, insulin resistance; MetS, metabolic syndrome; NASH, nonalcoholic steatohepatitis; RVH, renovascular hypertension; STZ, streptozotocin; TC, total cholesterol; TG, triglycerides; YL, Yorkshire × Landrace. Green, Presence or increase of parameter or disease state. Yellow, Modest or variable finding. Red, Unchanged parameter or absence of disease state. Blank, Not assessed or not reported. *Fig. 2 does not include the vascular and atherosclerotic consequences of MetS to avoid repetition with the content of other reviews (6).


Table 1. Representative parameters showing the spectrum of known porcine models of metabolic syndrome

Porcine Model of MetS Starting Age Study Duration, wk BW, kg SBP/DBP or MAP, mmHg FBG, mg/dL HOMA-IR TG, mg/dL TC, mg/dL HDL-C, mg/dL References
Livestock breeds
 Domestic na 8 MetS 31 111/54 16 196 40 (28)
Ctl 27 117/57 18 48 30
 Domestic 3 mo 16 MetS 93 146/110 109 1.9 15 372 144 (23)
Ctl 78 126/89 106 0.60 8.4 81 46
 Domestic + RVH 7 wo 16 MetS 87 130/99 154 12 446 136 (18)
Ctl 65 115/82 144 7.0 77 45
 Domestic + RVH na 16 MetS 51 149/115 6.0 425 159 (21, 37)
Ctl 49 125/97 7.5 92 105
 Iberian 2 to 3 yo 14 MetS 252 172/118 117 0.28 28 112 27 (54)
Ctl 208 148/95 113 0.15 21 68 31
 Duroc 9 to 10 wo 10 MetS 60 109 1.56 29 156 61 (38)
Ctl 53 81 1.09 22 126 45
 DLY 8 wo 13 MetS 70 116 42 102 37 (40)
Ctl 72 122 34 89 31
 Landrace + DOCA-HT 3 mo 12 MetS 74 165/97 81 3.3 66 553 209 (57, 58)
Ctl 67 102/58 62 1.0 30 69 38
 Yorkshire 8 wo 12 MetS 75 165 2.6 116 259 52 (123, 122)
Ctl 64 92 1.1 59 43 18
 Yorkshire 8 wo 12 MetS 40* 96 86 495 209 (116)
Ctl 30* 75 92 69 38
 YL + STZ + RVH 2 to 3 mo 26 MetS 79 115 409 0.4 103 650 197 (126)
Ctl 102 90 110 0.3 31 85 43
Miniature breeds
 Ossabaw na 16 MetS 69 128/87 85 6.2 67 486 75 (107, 106)
Ctl 47 111/73 74 3.9 43 87 40
 Ossabaw 5 to 10 mo 24 MetS 86 158/104 88 3.9 130 629 83 (79)
Ctl 57 110/62 78 2.0 24 71 39
 Ossabaw 5 to 6 wo 39 MetS 100 130/99 308 6.3 78 190 56 (100)
Ctl 37 110/72 121 0.91 28 80 41
 Ossabaw 7 mo 54 MetS 111 160/108 83 2.5 39 215 56 (94)
Ctl 64 116/77 67 0.8 25 48 18
 Ossabaw + AB-HT 2 mo 43 MetS 76 124/74 0.92 54 475 (88)
Ctl 46 89/64 0.65 24 75
 Göttingen 11 wo 43 MetS 54 72 112 657 49 (46)
Ctl 24 70 30 73 31
 Göttingen 6 to 7 mo 56 MetS 78 67 1.1 56 462 (43)
Ctl 39 62 0.47 30 66
 Göttingen 11 to 13 mo 70 MetS 115 70 5.5 35 90 (42)
Ctl 45 68 2 35 85
 Göttingen + DOCA-HT 14 mo 20 MetS 46 167/106 55 1.5 350 250 (47)
Ctl 32 95/69 50 1.2 75 25
 Lee-Sung 5 mo 26 MetS 70 150/85 80 45 100 65 (61)
Ctl 30 120/65 70 25 60 35
 Lee-Sung 2 yo 52 MetS 153 72 1.6 81 70 41 (60)
Ctl 98 65 0.2 17 70 30
 Microminipig 3 to 4 mo 60 MetS 58 146/na 130 9 232 59 (65)
Ctl 25 114/na 65 12 63 42
 Bama na 26 MetS 45 293 12 855 189 (9)
Ctl 30 89 2.4 134 65
 Bama 8 mo 26 MetS 58 100 5.9 70 145 87 (14)
Ctl 35 74 1.8 22 77 32
 Bama 6 mo 100 MetS 140 90 5.6 152 132 38 (10)
Ctl 51 119 1.5 39 46 10
 Yucatan 5 mo 26 MetS 79 108/52 51 0.70 43 95 (129)
Ctl 36 99/55 42 0.20 24 69
 Tibetan 3 mo 24 MetS 49 180/127 98 5.7 25 407 121 (133)
Ctl 33 127/90 84 3.7 24 96 34

AB-HT, aortic-banding-induced hypertension; BW, body weight; ctl, control group; DBP, diastolic blood pressure; DLY, duroc × landrace × yorkshire; DOCA-HT, deoxycorticosteroneactetate-induced hypertension; FBG, fasting blood glucose; HDL-C, high-density lipoprotein cholesterol; HOMA-IR, homeostatic model assessment for insulin resistance; MAP, mean arterial pressur; MetS, metabolic syndrome group; mo, months old; na, not assessed or not reported; RVH, renovascular hypertension; SBP, systolic blood pressure; STZ, streptozotocin; TC, total cholesterol; TG, triglycerides; wo, weeks old; YL, yorkshire × landrace; yo, years old.

*Body mass index (kg/m3).

Domestic Pig

Domestic pigs are large breeds mostly used as livestock. Although readily available and affordable, they present the disadvantage of rapidly increasing in size, thus complicating husbandry and handling over long-term interventions. It is of note that the designation “domestic pig” encompasses a large variety of breeds and crossbreeds that can differ in their anatomy and growth rate (134). Consequently, the domestic pig may suffer from poor standardization due to its ambiguous denomination.

The domestic pig exhibited all core features of MetS within 16 wk of “Western” diet intervention (25, 28, 33). This model developed moderate obesity, with 3-mo-old individuals reaching ∼90 kg of body weight after 16 wk of high-fat-fructose diet, and 70 kg when fed a standard diet. The high-fat-fructose diet intervention also consistently induced hypertension, insulin resistance, hypertriglyceridemia and hypercholesterolemia, as well as high high-density lipoprotein-cholesterol (HDL-C) levels. Interestingly, certain high-fat-cholesterol diets failed to induce obesity and hypertriglyceridemia to any convincing degree (17, 37). Finally, it is noteworthy that domestic pigs did not develop MetS over excessively short studies. Indeed, Shimabukuro et al. reported that an 8-wk long intervention solely produced hypercholesterolemia without evidence of concomitant hypertriglyceridemia, hypertension, or obesity in this model (28).

Although suffering from hypertension, high-fat-fructose diet-fed domestic pigs did not develop cardiac hypertrophy or contractile dysfunction (25, 28, 35). Authors only reported slight alterations of cardiac markers of metabolic and immune function (25, 28). These markers may nonetheless constitute useful tools for guiding disease diagnosis before the appearance of marked MetS comorbidities. Remarkably, the induction of renovascular hypertension (RVH) along with “Western” diet consistently produced cardiac structural changes with myocardial hypertrophy, fibrosis, inflammation, and impaired left ventricle (LV) function similar to those observed in patients suffering from HF-pEF within 16 wk of intervention (17, 18, 22, 37). Interestingly, RVH models had blood pressure parameters similar to diet-induced models at study termination, suggesting that the combination of RVH and “Western diet” did not aggravate hypertension. Therefore, the cardiac sequelae observed in these models may be attributable to an earlier induction of hypertension rather than a more severe phenotype. Finally, domestic pig fed high-fat-cholesterol or high-fat-fructose diet with renal artery coiling or stenosis accurately recapitulated the clinical features of renovascular disease (17, 32, 33).

The domestic pig stands out as a robust model of MetS with substantial cardiac remodeling upon surgically induced RVH. It is of note that dietary interventions alone did not produce marked secondary cardiac comorbidities, with a notable absence of myocardial hypertrophy, potentially due to time limitations inherent in the use of large porcine breeds. Finally, it is also important to consider that, due to their fast growth rate, such breeds as the domestic pig are generally studied over juvenile stages of development, a phenotype that differs from adult animals (135).

Iberian Pig

The Iberian pig is a breed of leptin-resistant animals reared in central and southern Spain which is predisposed to the development of intramuscular adiposity, obesity, and insulin resistance when maintained on a “Western”-type diet (55). Investigators should be aware of the large adult size of such animals, with lean Iberian pigs growing up to 200 kg within 2 yr, and typically exceeding 250 kg when fed a diet enriched in saturated fats (54). The Iberian model also develops spontaneous hypertension as a consequence of leptin resistance (136), with standard diet-fed animals presenting systolic and diastolic blood pressure of ∼150 and 95 mmHg, respectively. Remarkably, in a study conducted by Torres-Rovira et al. (54), Iberian pigs fed a diet rich in saturated fat developed MetS in only 3 mo, which is relatively short when compared with other models. In this study, animals had ad libitum access to food, which may have accelerated development of MetS. However, unrestricted food intake may also result in poor model reproducibility with increased inter-individual variability. Recently, Rodriguez et al. (53) characterized further the model by demonstrating secondary glomerular changes that were associated with the development of a “diabetoid” phenotype after 100 days of “high-fat diet”. Using the same breed, Hernandez et al. sought to develop a model of MetS with nonalcoholic steatohepatitis (NASH) in juvenile Iberian pigs fed a high-fat-fructose diet. Although NASH was successfully reproduced, the model failed to replicate certain core features of MetS including obesity and insulin resistance (56). This may have been caused by an excessively short study duration (10 wk) and the use of a piglet model (13 days old). In spite of this, authors demonstrated that the intervention resulted in robust alterations in gut microbiota, intestinal mucosa structure, and bile acid metabolism that may have initiated NASH and preceded the clinical manifestation of MetS (56).

Although the Iberian pig appears to be well suited to MetS research, previous studies on this model have produced conflicting results. Moreover, studies using this model remain scarce, which may be in part explained by the inherent commercial value of the Iberian pig (i.e., Jamón ibérico).

Landrace and Yorkshire Swine

The Landrace pig was originally bred in Denmark in the first half of the 20th century. The breed results from a cross between the Large White and the Danish native pig. The original Danish Landrace pig has been subsequently exported and crossed again, giving rise to diverse Landrace breeds (137). The Landrace swine has been selectively bred for intensive meat production and now represents one of the largest porcine models used in preclinical research. Landrace piglets gain over 100 kg in their first 6 mo; females reach ∼250 kg at maturity, whereas their male counterparts can weigh up to 320 kg. Landrace swine quickly develops organs with dimensions comparable to humans; however, an excessive body size complicates husbandry, pharmacological dosing, and experimental procedures. Consequently, the Landrace swine is generally studied during juvenile stages of development and over short periods. Finally, Landrace breed affordability represents a considerable asset in developing large animal models for preclinical research (137, 138).

Landrace swine fed a diet enriched in fat, salt, cholesterol, cholate, and sugar with mineralocorticoid (deoxycorticosterone acetate, DOCA)-induced hypertension developed MetS with HF-pEF within 12 wk of intervention. In this model, Schwarzl et al. (57) reported hypertriglyceridemia and hypercholesterolemia with secondary organ comorbidities including glomerular alterations and kidney fibrosis as well as cardiac hypertrophy and contractile dysfunction that were similar to that observed in the human HF-pEF. Interestingly, authors provided insights into the cardiovascular pathology by reporting a shift in titin isoforms that were indicative of myocardial stiffening. Our group furthered the characterization of this model for MetS core and secondary parameters by reporting glucose intolerance, insulin resistance, gut microbiota alteration, systemic inflammation, altered bile acid metabolism, and hepatic steatosis upon an identical intervention (58). Finally, Zhang et al. (59) induced HF-pEF by combining a “Western” diet with DOCA and angiotensin II-induced hypertension that resulted in marked cardiac hypertrophy and fibrosis as early as 9 wk after intervention. Authors also demonstrated that dapagliflozin, a sodium-glucose co-transporter 2 inhibitor, exerted a protective effect against MetS and HF-pEF, which supports the strength of the Landrace swine as a preclinical model.

The Yorkshire swine is one of the most common large commercial breeds in the world and is generally recognized for its lean mass and backfat development. Although this breed has found utility in studies concerned with vascular pathophysiology (115, 116, 123, 139), it has not been frequently employed in MetS studies. In fact, streptozotocin challenge was found to be necessary to induce true dysfunction in glucose metabolism for these animals in several studies (124, 125). Nonetheless, the Yorkshire breed may represent an affordable model with high relevance for MetS research since it effectively reproduced features of MetS organ sequelae with cardiac steatosis, fibrosis, inflammation, apoptosis and LV dysfunction; as well as hepatic steatosis upon a high-cholesterol diet (113, 115, 116). It is of note that the Yorkshire × Landrace crossbreed also represents a robust model of MetS secondary outcome with successful reproduction of microbiota alteration (127) and hepatic steatosis (125). Sorop et al. set out to develop a Yorkshire × Landrace model of MetS with secondary sequelae induced by streptozotocin injection, RVH, and high-fat-sugar-cholesterol diet. This model developed cardiac fibrosis, cardiomyocyte atrophy, and impaired LV contractile function after 6 mo of intervention (126).

The Landrace and Yorkshire livestock breeds represent useful models for studying MetS in larger, naturally leaner animals. Indeed, they have the notable advantage of being readily available in large numbers and can achieve most biochemical aspects of the MetS in a matter of months. The Landrace breed stands out as a more robust model, with already characterized secondary outcomes including microbiota, hepatic, and cardiovascular sequelae. Particularly, the combination of “Western” diet and DOCA repetitively demonstrated its reliability in inducing MetS with HF-pEF in this breed.

Ossabaw Minipig

The Ossabaw minipig is a small-sized animal that reaches 40 kg at sexual maturity. The breed is described in the literature as displaying a “thrifty” phenotype, with the ability to extract high proportions of energy from its diet and accrue fat mass with great efficiency (92). This animal was transferred from Spain to Ossabaw Island in Georgia, USA, as a future food source for the 16th century explorers. In the year 2000, in an attempt to protect the native loggerhead turtle population, the US Government ordered the eradication of Ossabaw breed from the island. Recognizing the potential value of such a pig in advancing MetS research, Dr. Michael Sturek of Indiana University School of Medicine led an expedition to capture and export 26 disease-free feral animals, with a view to setting up a breeding colony on the mainland (140). In an attempt to unravel Ossabaw’s phenotype, genetic investigation revealed a single amino acid mutation, Val199 to Ile, in the regulatory γ3 subunit of the AMP-activated kinase gene. This mutation is associated with reduced muscle glycogen and an increased level of intramuscular adiposity; two traits that are consistent with the Ossabaw “thrifty” phenotype and potentially key to its metabolic dysfunction (141). Finally, this “thrifty” phenotype closely resembles human physiology, which strengthens the relevance of the Ossabaw model for translational studies (142).

When maintained on a high-fat-fructose diet for 6 to 9 mo, the Ossabaw miniature swine developed every core feature of MetS (79, 94, 100, 106, 107). This model exhibited severe obesity and researchers reported that animals exceeded 100 kg over long-term interventions (94). In addition, Panasevich et al. (97, 102) demonstrated that a high-fat-fructose diet intervention induced intestinal permeability and alterations in gut microbe populations that were associated with immune system regulation and metabolic function. The Ossabaw minipig also stands out as a powerful model for cardiovascular research. Indeed, Olver et al. accurately recapitulated MetS-induced cardiovascular disease in Ossabaw miniature swine by inducing clinical features of HF-pEF using aortic banding on top of a high-fat-fructose diet over 16 mo (88). In this model, authors also investigated the pathological processes involved in right ventricular remodeling using transcriptomic analysis (109). Interestingly, Lee et al. (79, 83) compared the effects of three “Western” diets on MetS and NASH development in the Ossabaw model. These “fructose,” “atherogenic,” and “modified-atherogenic” diets were isocaloric and mainly differed in their sources of fat and carbohydrates. The “fructose” diet contained 72% carbohydrates and 10.5% fat, whereas the two “atherogenic” diets contained ∼40% carbohydrates and 45% fat. However, the “modified-atherogenic” diet was supplemented with coconut oil and lard on top of hydrogenated soybean oil. “Fructose” diet-fed animals only developed obesity and insulin resistance, whereas their “atherogenic” diet-fed counterparts accurately replicated MetS. Interestingly, the “modified-atherogenic” diet resulted in accentuated hypertriglyceridemia, hypercholesterolemia and resulted in soaring serum leptin and tumor necrosis factor-α levels. Finally, this “modified-atherogenic” diet also induced the full spectrum of NASH diagnosis criteria, whereas animals fed the “fructose” or “atherogenic” diet alone did not develop any of these disease features (83). These data highlight the importance of the source of dietary components when modeling MetS and NASH in the Ossabaw model. Differences between Ossabaw MetS models were also noticed when investigating adipose tissue (AT) inflammation. Vieira-Potter et al. (100) reported that Ossabaw miniature swine subjected to a “Western” diet over 9 mo developed MetS in absence of systemic and AT inflammation compared with low-fat diet-fed animals. Authors postulated that visceral AT inflammation was not an essential component of MetS pathophysiology in their Ossabaw model, potentially due to the breed’s “thrifty” phenotype that enabled AT expansion without inflammation. Supporting this concept, Uceda et al. (96) reported increased pericardial AT deposition in the absence inflammation following 26 wk of ad libitum high-fat-fructose diet. However, several independent studies conducted on the Ossabaw model have induced systemic and AT inflammation upon varied “Western” diets (83, 95, 99). These conflicting results imply that study parameters are critical for accurately reproducing the inflammatory aspect of MetS in Ossabaw miniature swine. The precise elements that dictate the development of systemic inflammation in this breed remain to be identified for improved model fidelity.

The Ossabaw minipig stands out as the most explored and characterized porcine model of MetS, with high relevance to human pathology. The robustness of this model is also supported by numerous interventional studies that have explored varied secondary comorbidities of MetS. Moreover, a recent study has produced a high-quality assembly of the Ossabaw genome, which will greatly facilitate gene-targeted experimentation in porcine models (142). However, the prolonged period of dietary intervention and the genetic background required for this model may increase studies cost and thereby limit the number of trials that can be conducted.

Göttingen Minipig

The Göttingen minipig was first explored as a potential model of MetS in 2001, when Johansen et al. (45) induced obesity and dyslipidemia, without any appreciable effect on insulin sensitivity, through a 5-wk long high-fat, high-energy diet intervention. More recent efforts to build upon this model by Christoffersen et al. (41) explored the relationship between sex and the development of MetS. The study showed that female Göttingen minipigs were prone to a more complete phenotype than their male counterparts, displaying increased adiposity, hypercholesterolemia, hypertriglyceridemia and, crucially, insulin resistance. The authors postulated that this may be explained by the fact that male Göttingen minipigs maintain a disproportionately high level of circulating testosterone and estradiol when compared with the females of the same breed, which may protect them from MetS. Further studies demonstrated that male and female Göttingen minipigs eventually develop insulin resistance after a 13 to 16 mo-long high-fat-fructose-cholesterol or high-fat, high-energy diet (42, 43, 46), suggesting again that the progression of MetS is slow when induced by a dietary intervention alone.

Van den Dorpel et al. (49) overcame this limitation by combining high-fat-cholesterol diet intervention with streptozotocin-induced β-cell toxicity that resulted in deregulated glucose and lipid metabolism with cardiac diastolic dysfunction as early as 3 mo after intervention. It is also of note that Göttingen minipigs developed remarkably severe obesity upon high-fat fructose cholesterol diet, with animals weighing up to 80 kg after 56 wk of intervention, whereas their standard diet-fed counterparts did not exceed 40 kg (43). More recently, Sharp et al. induced MetS with HF-pEF in Göttingen minipigs fed a high-fat-fructose-cholesterol diet with DOCA-induced hypertension. Whereas the model suffered from an excessive mineralocorticoid dosage, it effectively developed MetS core components with peripheral organ co-morbidities including clinical features of HF-pEF within 20 wk of intervention (47). Although the Göttingen model of MetS stands out as a suitable model for cardiovascular research, it hardly reproduces the hepatic sequelae of MetS. Indeed, although resulting in marked obesity and dyslipidemia, a 1-yr long high-fat-fructose-cholesterol diet intervention did not produce any observable NASH feature in this model (43). Finally, Renner et al. (42) reported that the Göttingen model fed a high-fat, high-energy diet for 70 wk exhibited visceral fat inflammation with fibrotic and necrotic lesions. Authors postulated that adipose tissue inflammation may have promoted insulin resistance in these animals.

The Göttingen minipig, and especially the female of this breed, may represent a particularly useful model for cardiovascular research in obesity and MetS. The relatively small size of this animal facilitates husbandry and pharmacological studies while allowing the use of human-scale imaging and therapeutics for cardiovascular disease.

Lee-Sung and Lanyu Minipigs

The Taiwanese Lee-Sung miniature breed is a Lee × Landrace cross, which grows to roughly the weight of an adult human in just over one-and-a-half year (143). The Lanyu minipig breed is a genetically distinct pig population which was discovered on the Lanyu Islet, off the coast of Taiwan (144). When given an “atherogenic” diet, the Lanyu presented a metabolically healthy obese phenotype, with increased body weight and adiposity but normal lipidemia and glycaemia (61), suggesting that the model might be best applied to diabetes research. On the other hand, the “atherogenic” diet-fed Lee-Sung minipig model accurately reproduced MetS with remarkably severe obesity after 6 mo of feeding. In the study performed by Li et al. (61), Lee-Sung minipigs fed a “Western” diet developed insulin resistance only after 6 mo of intervention, whereas hyperglycemia was detectable as early as 3 mo. In addition, this model developed hepatosteatosis with tissue fibrosis and inflammation, as well as myocardial hypertrophy, fibrosis, and steatosis (6062, 64).

Whereas the Lanyu minipig does not develop MetS, the Lee-Sung breed fed an “atherogenic” diet accurately reproduces every central feature of the disease along with hepatic and cardiac pathological changes.

Bama Minipig

Originating from Southern China, the Bama is a miniature pig purposely bred for meeting the needs of biomedical research (145). The Bama minipig is highly inbred, inexpensive, and maintains a small size at maturity with an average weight of 40 to 50 kg when maintained on a standard diet (10, 146). In addition, the Bama is a model of choice for infectious diseases and pharmacological studies (147). However, the application of Bama minipigs is limited on an international stage since it is a breed local to China and rarely found beyond borders of the country.

The Bama minipig fed a high-fat-fructose diet developed most core features of MetS with consistent induction of hypertriglyceridemia, hypercholesterolemia, and glucose intolerance (9, 10, 14). However, no evidence of hypertension has been reported in this model and, although Chen et al. (148) described the Bama breed as susceptible to developing type 2 diabetes, reports of hyperglycemia in this model remain contradictory (9, 10, 14). This miniature breed can reach 50 kg after 6 mo of high-fat-fructose diet and up to 140 kg after 2.5 yr of intervention (10). In addition, Xia et al. (10) reported that a 23-mo-long high-fat-fructose diet induced hepatic steatosis and inflammation. In the same model, authors reported cardiac sequelae with myocardial hypertrophy and steatosis as well as transcriptomic alterations in metabolic, apoptosis, and inflammatory signaling-related pathways (11). These results suggest that the Bama minipig may be a relevant translational model for NASH and cardiovascular research. Finally, this model also developed renal injury with lipid deposition, cellular apoptosis, tissue inflammation, and glomerular hypertrophy (15).

To date only mid- to long-term studies have been conducted on the Bama model, with durations ranging from 6 to 23 mo. Nonetheless, Zhu et al. (9) reported marked dyslipidemia, hyperglycemia, and hyperinsulinemia as early as 3 mo of high-fat-sucrose diet. Therefore, mid-term studies may produce satisfactory reproduction of MetS in the Bama model.

The Bama minipig stands out as a standardized and affordable breed of convenient size that emulates MetS core components and several of its secondary comorbidities, including nonalcoholic fatty liver disease and cardiac remodeling upon a “Western” diet.

Yucatan Minipig

The Yucatan minipig was imported from the Mexican peninsula of the same name in the 1960s (149). This hairless breed found use as an experimental model primarily due to its docile temperament and substantial tolerance to human handling and restraint. In a direct comparison with the Ossabaw miniature swine model, the Yucatan swine displayed a less severe phenotype with lower blood pressure, reduced body fat accumulation as well as more moderate glucose intolerance and insulin resistance (90). In spite of this, Huang et al. (129, 130) demonstrated the potential of Yucatan animals as a model of MetS, using a high-fat-fructose diet to induce hypertriglyceridemia, hypercholesterolemia, and insulin resistance and investigate contractile recovery after myocardial ischemia. However, this model developed only moderate hypertension with elevated systolic but unchanged diastolic blood pressure. Naturally, this high-fat-fructose diet-fed model did not exhibit severe myocardial sequelae, but instead only modest LV hypertrophy and unchanged contractile function. These results suggest that the diet-induced Yucatan MetS model poorly reproduces the cardiovascular consequences of MetS. Marshall et al. (132) demonstrated that this model can be improved by artificially inducing MetS components such as hypertension when they replicated HF-pEF in aortic-banded Yucatan minipigs. Interestingly, LV hypertrophy was detectable in this model as early as 2 mo after banding. More recently, Curtasu et al. (128) performed a comparison between ad libitum and restricted feeding patterns with a high-fat-sugar diet. Yucatan minipigs with restricted food access gained ∼30 kg in a 20 wk intervention, whereas animals fed ad libitum gained over 50 kg. Ad libitum feeding pattern also resulted in more severe hypercholesterolemia; however, serum triglycerides and glucose levels did not differ between groups. Finally, Pop et al. (131) used the Yucatan model to examine gut hormone signaling in MetS following embolization of gastroduodenal arteries. This procedure was successful in attenuating ghrelin secretion and substantially altered expression of key glucose transporters, sodium-glucose-linked transporter-1, and glucose transporter-2, in the porcine foregut mucosa.

Overall, each core feature of MetS has been achieved in varying combinations in the different porcine models presented herein. A dietary intervention with variable feeding patterns was applied for inducing MetS in each porcine model discussed. These diets were generally categorized as “Western”-type, with increased fat, carbohydrate, cholesterol, and salt content to promote hypertension and metabolic dysfunction. However, although most “Western” diets were similar, their precise composition varied across studies. On top of this, diets also differed in the quantity of food that was distributed to animals. Researchers must consider that nutrient source and feeding pattern have been shown to considerably influence MetS progression (83, 150). Consistent design of dietary interventions would improve porcine model reproducibility.

The chemical or surgical induction of hypertension in combination with the “Western” diet intervention reliably replicated the cardiovascular sequelae of MetS and HF-pEF, such as cardiac hypertrophy, fibrosis, and LV contractile dysfunction in several models. By artificially promoting certain components of MetS, researchers can accelerate disease progression, which facilitates the replication of advanced MetS comorbidities within the allocated study duration. However, although requiring increased amounts of resources, long-term studies may more accurately replicate the long-term “natural” progression of the human MetS (6).

CHARACTERISTICS, ADVANTAGES, AND LIMITATIONS OF PORCINE MODELS OF METABOLIC SYNDROME AND EMERGING AREAS OF INVESTIGATION

Overall, porcine models fall into two major categories: livestock breeds and miniature breeds. Livestock breeds are large and lean animals that have been purposely selected for building up muscle mass. These breeds are affordable and available; however, their excessive size often necessitates the use of juvenile animals over short-term interventions. In addition, livestock species are generally outbred with a certain degree of heterozygosity and inter-individual genetic variability (151). Outbred animals better approximate the human diversity than homozygous inbred models (152); however, increased inter-individual variability implicates the use of larger cohorts to detect significant experimental outcomes.

On the other hand, miniature breeds are small and more suited to biomedical research, several of them such as the Ossabaw and Göttingen being naturally predisposed to obesity. Although coming at increased cost, these breeds have the major advantage of greatly facilitating long-term experiments, which can more reliably induce MetS core and secondary components.

In a recent review, Sturek et al. have extensively compared the Yucatan, Ossabaw, and Göttingen miniature models for their fidelity in reproducing MetS, type 2 diabetes, and atherosclerosis, as well as their respective contribution to cardiovascular device development (6). Since numerous other breeds of the Sus scrofa species have been used to emulate and explore MetS pathophysiology, we propose extending the comparison by including every model that has been characterized in any detail to date. Although highly similar, each porcine model presents specific advantages and limitations that are inherent to the breed itself and to the specific disease induction methods. It is also important to consider that porcine models differ in their MetS baseline parameters, such as blood pressure and body weight. Therefore, disease characterization is breed-specific and must be performed by comparing MetS versus lean animals instead of referring to preestablished threshold values as in humans. Here we collected representative MetS parameters for each porcine model to facilitate model comparison and characterization (Table 1). Moreover, we also propose consideration of MetS pathology with its cardiac and hepatic comorbidities (Fig. 2), as well as discussing emerging concepts such as gut microbiota alteration, intestinal permeability, and neurological disorders.

Metabolic Syndrome Core Components

Truncal obesity, hypertension, hypertriglyceridemia hypercholesterolemia, type 2 diabetes, and insulin resistance constitute the core components of MetS (1). The reliable induction of these parameters is essential for an accurate reproduction of the disease. Importantly, core parameter severity can influence the development of secondary organ sequelae and therefore the relevance of certain porcine models for specific aspects of MetS. In this section, we discuss the notable differences between porcine models and between porcine and human MetS.

Obesity.

Although every porcine model reliably reproduced the obesity component of MetS, differences in breeds’ phenotype were found to affect disease progression. Livestock breeds such as the domestic, Iberian, Landrace, Duroc, and the Yorkshire tended to exhibit a leaner phenotype than miniature breeds. Indeed, these MetS models generally exhibited moderate obesity, with a 10% to 30% increase in body weight compared with lean animals (Table 1). This may be explained by years of selective breeding of livestock animals in favor of muscle growth rather than fat accumulation. Meanwhile, miniature breeds including the Bama, Gottingen, Lee-Sung, Ossabaw, and Yucatan model of MetS developed severe obesity by building up to twice the body weight of control animals.

Hypertension.

Hypertension has been successfully induced in most porcine models of MetS via “Western” diets alone or in combination with pharmaceutical or surgical procedures. Although the normotensive and hypertensive parameters of most porcine MetS models closely resembled human criteria, the Iberian (53, 54) and Tibetan (133) models exhibited higher arterial pressures than other breeds, and Yucatan minipigs presented low diastolic blood pressure (129). These differences have to be considered when defining MetS in these models; this may also affect the severity of secondary comorbidities, especially cardiac sequelae.

Hypertriglyceridemia and hypercholesterolemia.

Although hypertriglyceridemia and hypercholesterolemia have been systematically reproduced in every porcine MetS model, it is important to consider that porcine HDL-C levels exhibit a pattern that is opposite to the human syndrome. Whereas low HDL-C constitute a MetS criteria in patients, porcine models of MetS exhibit higher HDL-C levels than their lean counterparts. This may be explained by the fact that MetS is generally induced with diets rich in cholesterol that results in severe hypercholesterolemia, with increased content in every lipoprotein fraction.

Type 2 diabetes and insulin resistance.

It is important to consider that, although most porcine models of MetS presented glucose intolerance and insulin resistance, they did not reliably exhibit type 2 diabetes with increased fasting blood glucose upon a dietary intervention. This may be due to the fact that pigs have increased pancreatic β cell capacity, resulting in improved glycemic control, which has been promoted over years of selective breeding for improved growth and thus energy accumulation (153). The use of juvenile animals may also have accounted for the poor reproduction of diabetes; indeed, young pigs have better glycemic control than adults (72). Consequently, the reproduction of a diabetic phenotype in porcine models generally required the induction of pancreatic dysfunction using toxic compounds such as streptozotocin or alloxan (43, 154). However, since the chemical induction of diabetes also impedes energy storage, it may result in an inaccurate reproduction of the obesity component of MetS (155). Most importantly, streptozotocin directly affects hepatic (156), cardiac (157), and renal function (158), and therefore influences the development of MetS secondary comorbidities. Investigators must also consider that pigs are resistant to streptozotocin toxicity, with no visible effect at low dose (50 mg/kg) and a partially reversible diabetic phenotype at higher dose (150 mg/kg) (159). Nonetheless, Hara et al. (160) reported that a 200 mg/kg dose induced stable diabetes over 20 wk without renal and hepatic dysfunction in Large White sows. However, a high dose of streptozotocin resulted in the rapid induction of pancreatic dysfunction that approximated the insulin-dependent type 1 diabetes phenotype (161).

Nonalcoholic Steatohepatitis

The most accurate reproductions of NASH were obtained in the Ossabaw miniature swine (83, 97) and Microminipig (65) (Table 2). Although both models reflected the full disease complexity, the Ossabaw fidelity was supported by independent studies, whereas the Microminipig has only recently started to emerge. The Göttingen (43, 50), Iberian (56), Lee-Sung (61, 64,) and Bama (10, 12, 13) remain satisfactory models that recapitulated most features of NASH. Intriguingly, different studies using the same breed did not consistently report similar NASH features. These discrepancies may be explained by differences between study parameters and design such as diet composition and intervention length. Identifying the parameters that are key for the induction of MetS comorbidities would improve the fidelity of porcine models as well as our grasp of disease pathogenesis.

Table 2. Reproduction of nonalcoholic steatohepatitis in porcine models of metabolic syndrome

NASH Criterion Microminipig Ossabaw Göttingen Iberian Lee-Sung Bama
Steatosis Yes (65) Yes (83, 97)
No (84, 101)
Yes (42)
No (43)
Yes (56) Yes (61, 64) Yes (10, 13)
Inflammation Yes (65, 66) Yes (83, 97)
No (84)
Yes (43) Yes (56) Yes (64) Yes (10)
Ballooning Yes (65) Yes (83, 84, 97) No (43) Yes (56) na na
Fibrosis Yes (65) Yes (83, 84, 88, 97) Yes (43, 47) No (56) Yes (64) na

na, not assessed or not reported; NASH, nonalcoholic steatohepatitis.

Cardiac Remodeling and Pathological Signaling in the Heart

When subjected to a “Western” diet challenge, most porcine models of MetS did not reliably develop features of cardiac remodeling. It is arguable that diet-induced models of MetS only reflected the early pathological changes that preceded the manifestation of cardiovascular sequelae; therefore, more pronounced comorbidities would have required longer interventions to develop. In line with this, the Bama minipig fed high-fat-sucrose diet for an extended period of 23 mo effectively developed severe cardiac hypertrophy with evidence of fibrosis and steatosis (10, 11). Although authors did not assess LV function, this study may constitute the only satisfactory reproduction of myocardial sequelae in a porcine model of MetS subjected to a dietary intervention alone. Therefore, the induction of additional components of MetS appears to be essential for accurately reproducing advanced states of cardiovascular diseases within short intervention periods. Indeed, the surgical or pharmaceutical induction of hypertension in domestic (17, 22, 37), Göttingen (47), Landrace (5759) and Ossabaw (88) models of MetS successfully recapitulated the cardiovascular secondary comorbidities of MetS. Importantly, these models exhibited pathological features that were analogous to human HF-pEF, which demonstrated the translatability of these porcine models in cardiovascular research. Therefore, the induction of hypertension in addition to a dietary challenge stands out as the most robust method for reproducing the complexity of structural heart diseases in varied porcine MetS models. These findings closely mirror human pathology where the mechanical stress induced by hypertension appears to contribute greatly to cardiac remodeling (162). Finally, although the myocardial sequelae of MetS have been shown to promote an atrial fibrillation substrate (163), this aspect remains unexplored in porcine models of MetS.

Emerging Concepts of Gut Microbiota Alteration, Intestinal Permeability, and Neurological Disorders

Recent research suggests the involvement of intestinal barrier disruption and gut mucosal inflammation in MetS pathophysiology (164). In line with this, the gut microbiota constitutes a major source of environmental factors and may be a key regulator of MetS progression. Indeed, several large-scale clinical studies have now demonstrated the metabolically influential role of microbiome-derived metabolites, such as branched-chain fatty acids (165), short-chain fatty acids (166), and modified bile acids (167). With dietary habits and digestive tract anatomy that are highly similar to humans, pigs represent excellent translational models for gastrointestinal research (5). Pigs are also excellent responders to dietary interventions as suggested by several studies reporting improvements in intestinal barrier function following prebiotic and probiotic supplementation (168170). Indeed, porcine MetS models exhibited alterations in gut microbiota that were consistent with those observed in human patients (51, 56, 58, 97, 102). Importantly, authors reported changes in key influential bacterial species that are involved in host metabolism, inflammatory-signaling, and bile acid modification (171). However, it is of note that although pigs and humans host similar bacterial populations, Bifidobacterium genera were found in lower abundance in the porcine gut microbiome (172). In addition, microbiota composition varied between porcine breeds and models. For instance, Pedersen et al. (51) reported that, compared with Göttingen minipigs, the gut microbiota of Ossabaw miniature swine was richer in Actinobacteria and Bacteroides. Interestingly, white pigs such as the Landrace have been shown to host more abundant cellulolytic communities than black-colored breeds (173).

In regard to intestinal homeostasis, only the Ossabaw (97, 98) and Iberian (56) models were examined for pathological changes in the gut mucosa during MetS. In these models, authors reported structural, cellular, and molecular intestinal changes that were consistent with decreased barrier function. However, no study so far has performed in vivo or ex vivo assessments of intestinal permeability in a porcine MetS model. The fidelity of porcine models in reproducing changes in intestinal homeostasis during MetS should be further explored.

Remarkably, the alterations in the gut microbiota that occur during MetS have been associated with neuroinflammation and neuroendocrine system deregulation during MetS (174). In a Duroc pig model of obesity, Valent et al. (39) evaluated the effects of the probiotic Bifidobacterium breve and omega-3 fatty acids over hypothalamic neurotransmitters. In this study, pigs fed a diet enriched in fat presented alterations in hypothalamic and hippocampal dopaminergic neurotransmitters that were reversed upon supplementation of B. breve and omega-3 fatty acids to diet. In addition, Olver et al. investigated the neurological sequelae of MetS and HF-pEF in the aortic-banded “Western” diet-fed Ossabaw minipig model. Authors reported impaired brain blood supply and alterations in cerebral vascular endothelium that were associated with transcriptomic signatures of neurological and behavioral disorders, including dementia and Alzheimer’s disease (175). Interestingly, cerebral vasoconstriction and reduced blood flow were attributed to impaired insulin-dependent vasodilatation (175, 176). These data strongly encourage the use of porcine models for investigating the neurological sequelae of MetS.

Overall, the swine stands out as a powerful model for studying MetS development with multiple end-organ sequelae. The strength of this model mainly lies in its similarities to human physiology and its accuracy in reproducing disease phenotype. However, some differences between porcine and human MetS have to be considered, the major one being the pig’s natural resistance to diabetes. Finally, researchers have to bear in mind that this large animal model comes with size and breed-related limitations (Table 3).

Table 3. Major strengths and limitations of the porcine models of metabolic syndrome

Strengths Limitations
  • Pig size allows large sample collection and human-tailored interventions

  • Gastrointestinal, immune, and cardiovascular systems similar to human

  • Livestock breeds are cost-efficient and readily available

  • Miniature breeds can be studied over long-term interventions

  • Pig size implies costly husbandry needs as well as specific equipment and expertise

  • The pig is a suboptimal model for diabetes

  • Large breeds generally studied over juvenile stages only

  • Certain models remain poorly standardized with conflicting studies

STRENGTHS AND LIMITATIONS

This review has applied a comprehensive search strategy that has outlined in detail the metabolic features reported in 127 articles and synthesized a condensed view of each distinct porcine model available. However, it is important to note that the authors may not have classified their model as MetS, but rather by the individual risk factors (i.e., obese, insulin resistant, hypertensive, etc.). The result of this potential misclassification could be that studies may have been erroneously excluded or missed during the initial search phase. In line with this, an additional informal review of the literature conducted using “pig model” and “risk factors” as search keywords revealed several additional porcine models of metabolic dysfunction. Although this uncovered several additional important citations, it is possible that other relevant studies have been omitted. Despite this limitation, this review has covered in detail the most complete and commonly applied porcine models of MetS.

CONCLUDING REMARKS

Herein, we have identified the individual characteristics of MetS that are associated with each porcine model, thereby providing researchers in the gastrointestinal and cardiometabolic fields with a comprehensive reference material from which to select the model most appropriate to their study design and objectives. In addition, this review indicates that there is currently no single model that perfectly recapitulates the human syndrome. Nevertheless, several porcine models pose highly satisfactory representations of MetS for translational research. The Ossabaw miniature swine is the most commonly used porcine MetS model and it also represents the most promising recapitulation of MetS due to its high fidelity in reproducing NASH and HF-pEF. The “Western” diet and mineralocorticoid-induced Landrace model represents a more affordable alternative that successfully reproduces intestinal and cardiovascular MetS comorbidities. Finally, miniature breeds such as the Göttingen and Bama allow researchers to investigate the progression of MetS over long-term studies. Future efforts should focus on further optimizing and characterizing currently available MetS models, while exploring adjacent parameters, such as the composition of the gut microbiome and development of the important extrametabolic sequelae commonly observed in patients with MetS.

GRANTS

This work was supported by Science Foundation of Ireland Grant R18769.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

G.L.C., P.M.R., F.M.H., and N.M.C. conceived and designed research; G.L.C., P.M.R., F.M.H., and N.M.C. prepared figures; G.L.C., P.M.R., and F.M.H. drafted manuscript; G.L.C., P.M.R., F.M.H., and N.M.C. edited and revised manuscript; N.M.C. approved final version of manuscript.

REFERENCES

  • 1. Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 20: 12 , 2018. doi:10.1007/s11906-018-0812-z.
    Crossref | PubMed | ISI | Google Scholar
  • 2. Mechanick JI, Farkouh ME, Newman JD, Garvey WT. Cardiometabolic-based chronic disease, addressing knowledge and clinical practice gaps. J Am Coll Cardiol 75: 539–555, 2020. doi:10.1016/j.jacc.2019.11.046.
    Crossref | PubMed | ISI | Google Scholar
  • 3. NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet 390: 2627–2642, 2017. doi:10.1016/S0140-6736(17)32129-3.
    Crossref | PubMed | ISI | Google Scholar
  • 4. He W, Goodkind D, Kowal P. An aging world: 2015. Int Population Rep 16: 95 , 2016. http://www.census.gov/library/publications/2016/demo/P95-16-1.html.
    Google Scholar
  • 5. Heinritz SN, Mosenthin R, Weiss E. Use of pigs as a potential model for research into dietary modulation of the human gut microbiota. Nutr Res Rev 26: 191–209, 2013. doi:10.1017/S0954422413000152.
    Crossref | PubMed | ISI | Google Scholar
  • 6. Sturek M, Alloosh M, Sellke FW. Swine disease models for optimal vascular engineering. Annu Rev Biomed Eng 22: 25–49, 2020. doi:10.1146/annurev-bioeng-082919-053009.
    Crossref | PubMed | ISI | Google Scholar
  • 7. Wang D, Xu X, Zhao M, Wang X. Accelerated miniature swine models of advanced atherosclerosis: a review based on morphology. Cardiovasc Pathol 49: 107241 , 2020. doi:10.1016/j.carpath.2020.107241.
    Crossref | PubMed | ISI | Google Scholar
  • 8. Hamamdzic D, Wilensky RL. Porcine models of accelerated coronary atherosclerosis: role of diabetes mellitus and hypercholesterolemia. J Diabetes Res 2013: 761415 , 2013. doi:10.1155/2013/761415.
    Crossref | PubMed | ISI | Google Scholar
  • 9. Zhu X, Zhang H, Chen HN, Deng XJ, Tu Y, Jackson AO, Qing JN, Wang A, Patel V, Yin K. Perivascular adipose tissue dysfunction aggravates adventitial remodeling in obese mini pigs via NLRP3 inflammasome/IL-1 signaling pathway. Acta Pharmacol Sin 40: 46–54, 2019. doi:10.1038/s41401-018-0068-9.
    Crossref | PubMed | ISI | Google Scholar
  • 10. Xia J, Yuan J, Xin L, Zhang Y, Kong S, Chen Y, Yang S, Li K. Transcriptome analysis on the inflammatory cell infiltration of nonalcoholic steatohepatitis in Bama minipigs induced by a long-term high-fat, high-sucrose diet. PLoS One 9: e113724 , 2014. doi:10.1371/journal.pone.0113724.
    Crossref | PubMed | ISI | Google Scholar
  • 11. Xia J, Zhang Y, Xin L, Kong S, Chen Y, Yang S, Li K. Global transcriptomic profiling of cardiac hypertrophy and fatty heart induced by long-term high-energy diet in Bama miniature pigs. PLoS One 10: e0132420 , 2015. doi:10.1371/journal.pone.0132420.
    Crossref | PubMed | ISI | Google Scholar
  • 12. Xia J, Xin L, Zhu W, Li L, Li C, Wang Y, Mu Y, Yang S, Li K. Characterization of long non-coding RNA transcriptome in high-energy diet induced nonalcoholic steatohepatitis minipigs. Sci Rep 6: 30709 , 2016. doi:10.1038/srep30709.
    Crossref | PubMed | ISI | Google Scholar
  • 13. Yang SL, Xia JH, Zhang YY, Fan JG, Wang H, Yuan J, Zhao ZZ, Pan Q, Mu YL, Xin LL, Chen YX, Li K. Hyperinsulinemia shifted energy supply from glucose to ketone bodies in early nonalcoholic steatohepatitis from high-fat high-sucrose diet induced Bama minipigs. Sci Rep 5: 13980 , 2015. doi:10.1038/srep13980.
    Crossref | PubMed | ISI | Google Scholar
  • 14. Liu Y, Yuan J, Xiang L, Zhao Y, Niu M, Dai X, Chen H. A high sucrose and high fat diet induced the development of insulin resistance in the skeletal muscle of Bama miniature pigs through the Akt/GLUT4 pathway. Exp Anim 66: 387–395, 2017. doi:10.1538/expanim.17-0010.
    Crossref | PubMed | ISI | Google Scholar
  • 15. Li L, Zhao Z, Xia J, Xin L, Chen Y, Yang S, Li K. A long-term high-fat/high-sucrose diet promotes kidney lipid deposition and causes apoptosis and glomerular hypertrophy in Bama minipigs. PLoS One 10: e0142884 , 2015. doi:10.1371/journal.pone.0142884.
    Crossref | PubMed | ISI | Google Scholar
  • 16. Yang B, Xu B, Zhao H, Wang YB, Zhang J, Li CW, Wu Q, Cao YK, Li Y, Cao F. Dioscin protects against coronary heart disease by reducing oxidative stress and inflammation via Sirt1/Nrf2 and p38 MAPK pathways. Mol Med Rep 18: 973–980, 2018. doi:10.3892/mmr.2018.9024.
    Crossref | PubMed | ISI | Google Scholar
  • 17. Urbieta-Caceres VH, Zhu XY, Jordan KL, Tang H, Textor K, Lerman A, Lerman LO. Selective improvement in renal function preserved remote myocardial microvascular integrity and architecture in experimental renovascular disease. Atherosclerosis 221: 350–358, 2012. doi:10.1016/j.atherosclerosis.2011.10.005.
    Crossref | PubMed | ISI | Google Scholar
  • 18. Go JL, Prem K, Al-Hijji MA, Qin Q, Noble C, Young MD, Lerman LO, Lerman A. Experimental metabolic syndrome model associated with mechanical and structural degenerative changes of the aortic valve. Sci Rep 8: 17835 , 2018. doi:10.1038/s41598-018-36388-y.
    Crossref | PubMed | ISI | Google Scholar
  • 19. Eirin A, Hedayat AF, Ferguson CM, Textor SC, Lerman A, Lerman LO. Mitoprotection preserves the renal vasculature in porcine metabolic syndrome. Exp Physiol 103: 1020–1029, 2018. doi:10.1113/EP086988.
    Crossref | PubMed | ISI | Google Scholar
  • 20. Meng Y, Eirin A, Zhu XY, Tang H, Hickson LJ, Lerman A, van Wijnen AJ, Lerman LO. Micro-RNAS regulate metabolic syndrome-induced senescence in porcine adipose tissue-derived mesenchymal stem cells through the P16/MAPK pathway. Cell Transplant 27: 1495–1503, 2018. doi:10.1177/0963689718795692.
    Crossref | PubMed | ISI | Google Scholar
  • 21. Eirin A, Zhu XY, Urbieta-Caceres VH, Grande JP, Lerman A, Textor SC, Lerman LO. Persistent kidney dysfunction in swine renal artery stenosis correlates with outer cortical microvascular remodeling. Am J Physiol Renal Physiol 300: F1394–F1401, 2011. doi:10.1152/ajprenal.00697.2010.
    Link | ISI | Google Scholar
  • 22. Eirin A, Williams BJ, Ebrahimi B, Zhang X, Crane JA, Lerman A, Textor SC, Lerman LO. Mitochondrial targeted peptides attenuate residual myocardial damage after reversal of experimental renovascular hypertension. J Hypertens 32: 154–165, 2014. doi:10.1097/HJH.0b013e3283658a53.
    Crossref | PubMed | ISI | Google Scholar
  • 23. Meng Y, Eirin A, Zhu XY, Tang H, Chanana P, Lerman A, Van Wijnen AJ, Lerman LO. The metabolic syndrome alters the miRNA signature of porcine adipose tissue-derived mesenchymal stem cells. Cytometry A 93: 93–103, 2018. doi:10.1002/cyto.a.23165.
    Crossref | PubMed | ISI | Google Scholar
  • 24. Eirin A, Woollard JR, Ferguson CM, Jordan KL, Tang H, Textor SC, Lerman A, Lerman LO. The metabolic syndrome induces early changes in the swine renal medullary mitochondria. Transl Res 184: 45–56.e9, 2017. doi:10.1016/j.trsl.2017.03.002.
    Crossref | PubMed | ISI | Google Scholar
  • 25. Yuan F, Woollard JR, Jordan KL, Lerman A, Lerman LO, Eirin A. Mitochondrial targeted peptides preserve mitochondrial organization and decrease reversible myocardial changes in early swine metabolic syndrome. Cardiovasc Res 114: 431–442, 2018. doi:10.1093/cvr/cvx245.
    Crossref | PubMed | ISI | Google Scholar
  • 26. Nargesi AA, Zhang L, Tang H, Jordan KL, Saadiq IM, Textor SC, Lerman LO, Eirin A. Coexisting renal artery stenosis and metabolic syndrome magnifies mitochondrial damage, aggravating poststenotic kidney injury in pigs. J Hypertens 37: 2061–2073, 2019. doi:10.1097/HJH.0000000000002129.
    Crossref | PubMed | ISI | Google Scholar
  • 27. Zhang LH, Zhu XY, Eirin A, Nargesi AA, Woollard JR, Santelli A, Sun IO, Textor SC, Lerman LO. Early podocyte injury and elevated levels of urinary podocyte-derived extracellular vesicles in swine with metabolic syndrome: role of podocyte mitochondria. Am J Physiol Renal Physiol 317: F12–F22, 2019. doi:10.1152/ajprenal.00399.2018.
    Link | ISI | Google Scholar
  • 28. Shimabukuro M, Okawa C, Yamada H, Yanagi S, Uematsu E, Sugasawa N, Kurobe H, Hirata Y, Kim-Kaneyama J, Lei X-F, Takao S, Tanaka Y, Fukuda D, Yagi S, Soeki T, Kitagawa T, Masuzaki H, Sato M, Sata M. The pathophysiological role of oxidized cholesterols in epicardial fat accumulation and cardiac dysfunction: a study in swine fed a high caloric diet with an inhibitor of intestinal cholesterol absorption, ezetimibe. J Nutr Biochem 35: 66–73, 2016. doi:10.1016/j.jnutbio.2016.05.010.
    Crossref | PubMed | ISI | Google Scholar
  • 29. Nargesi AA, Zhu XY, Conley SM, Woollard JR, Saadiq IM, Lerman LO, Eirin A. Renovascular disease induces mitochondrial damage in swine scattered tubular cells. Am J Physiol Renal Physiol 317: F1142–F1153, 2019. doi:10.1152/ajprenal.00276.2019.
    Link | ISI | Google Scholar
  • 30. Ferguson CM, Eirin A, Michalak GJ, Hedayat AF, Abumoawad A, Saad A, Zhu X, Textor SC, McCollough CH, Lerman LO. Intrarenal fat deposition does not interfere with the measurement of single-kidney perfusion in obese swine using multi-detector computed tomography. J Cardiovasc Comput Tomogr 12: 149–152, 2018. doi:10.1016/j.jcct.2018.01.003.
    Crossref | PubMed | ISI | Google Scholar
  • 31. Farahani RA, Afarideh M, Zhu XY, Tang H, Jordan KL, Saadiq IM, Ferguson CM, Lerman A, Textor SC, Lerman LO, Eirin A. Percutaneous transluminal renal angioplasty attenuates poststenotic kidney mitochondrial damage in pigs with renal artery stenosis and metabolic syndrome. J Cell Physiol 236: 4036–4049, 2021. doi:10.1002/jcp.30146.
    Crossref | PubMed | ISI | Google Scholar
  • 32. Chade AR, Williams ML, Engel J, Guise E, Harvey TW. A translational model of chronic kidney disease in swine. Am J Physiol Renal Physiol 315: F364–F373, 2018. doi:10.1152/ajprenal.00063.2018.
    Link | ISI | Google Scholar
  • 33. Pawar AS, Zhu XY, Eirin A, Tang H, Jordan KL, Woollard JR, Lerman A, Lerman LO. Adipose tissue remodeling in a novel domestic porcine model of diet-induced obesity. Obesity (Silver Spring) 23: 399–407, 2015. doi:10.1002/oby.20971.
    Crossref | PubMed | ISI | Google Scholar
  • 34. Eirin A, Zhu XY, Woollard JR, Tang H, Dasari S, Lerman A, Lerman LO. Metabolic syndrome interferes with packaging of proteins within porcine mesenchymal stem cell-derived extracellular vesicles. Stem Cells Transl Med 8: 430–440, 2019. doi:10.1002/sctm.18-0171.
    Crossref | PubMed | ISI | Google Scholar
  • 35. Meng Y, Eirin A, Zhu XY, O'Brien DR, Lerman A, van Wijnen AJ, Lerman LO. The metabolic syndrome modifies the mRNA expression profile of extracellular vesicles derived from porcine mesenchymal stem cells. Diabetol Metab Syndr 10: 58 , 2018. doi:10.1186/s13098-018-0359-9.
    Crossref | PubMed | ISI | Google Scholar
  • 36. Zhang X, Kim SR, Ferguson CM, Ebrahimi B, Hedayat AF, Lerman A, Lerman LO. The metabolic syndrome does not affect development of collateral circulation in the poststenotic swine kidney. Am J Hypertens 31: 1307–1316, 2018. doi:10.1093/ajh/hpy127.
    Crossref | PubMed | ISI | Google Scholar
  • 37. Eirin A, Zhu XY, Ferguson CM, Riester SM, Van Wijnen AJ, Lerman A, Lerman LO. Intra-renal delivery of mesenchymal stem cells attenuates myocardial injury after reversal of hypertension in porcine renovascular disease. Stem Cell Res Ther 6: 7 , 2015. doi:10.1186/scrt541.
    Crossref | PubMed | ISI | Google Scholar
  • 38. Jové M, Tibau J, Serrano JC, Berdún R, Rodríguez-Palmero M, Font-I-Furnols M, Cassanyé A, Rodriguez-Mortera R, Sol J, Rassendren H, Fàbrega E, Crescenti A, Castell A, Sabater M, Ortega FJ, Martin-Gari M, Quintanilla R, Puigjaner J, Moreno JA, Prat J, Arola L, Fernández-Real JM, Pamplona R, Portero-Otin M. Molecular phenomics of a high-calorie diet-induced porcine model of prepubertal obesity. J Nutr Biochem 83: 108393 , 2020. doi:10.1016/j.jnutbio.2020.108393.
    Crossref | PubMed | ISI | Google Scholar
  • 39. Valent D, Arroyo L, Fàbrega E, Font-I-Furnols M, Rodríguez-Palmero M, Moreno-Muñoz JA, Tibau J, Bassols A. Effects of a high-fat-diet supplemented with probiotics and ω3-fatty acids on appetite regulatory neuropeptides and neurotransmitters in a pig model. Benef Microbes 11: 347–359, 2020. doi:10.3920/BM2019.0197.
    Crossref | PubMed | ISI | Google Scholar
  • 40. Yang XF, Qiu YQ, Wang L, Gao KG, Jiang ZY. A high-fat diet increases body fat mass and up-regulates expression of genes related to adipogenesis and inflammation in a genetically lean pig. J Zhejiang Univ Sci B 19: 884–894, 2018. doi:10.1631/jzus.B1700507.
    Crossref | PubMed | ISI | Google Scholar
  • 41. Christoffersen B, Golozoubova V, Pacini G, Svendsen O, Raun K. The young Göttingen minipig as a model of childhood and adolescent obesity: influence of diet and gender. Obesity (Silver Spring) 21: 149–158, 2013. doi:10.1002/oby.20249.
    Crossref | PubMed | ISI | Google Scholar
  • 42. Renner S, Blutke A, Dobenecker B, Dhom G, Müller TD, Finan B, Clemmensen C, Bernau M, Novak I, Rathkolb B, Senf S, Zöls S, Roth M, Götz A, Hofmann SM, Hrabĕ de Angelis M, Wanke R, Kienzle E, Scholz AM, DiMarchi R, Ritzmann M, Tschöp MH, Wolf E. Metabolic syndrome and extensive adipose tissue inflammation in morbidly obese Göttingen minipigs. Mol Metab 16: 180–190, 2018. doi:10.1016/j.molmet.2018.06.015.
    Crossref | PubMed | ISI | Google Scholar
  • 43. Schumacher-Petersen C, Christoffersen BO, Kirk RK, Ludvigsen TP, Zois NE, Pedersen HD, Vyberg M, Olsen LH. Experimental non-alcoholic steatohepatitis in Göttingen minipigs: consequences of high fat-fructose-cholesterol diet and diabetes. J Transl Med 17: 110 , 2019. doi:10.1186/s12967-019-1854-y.
    Crossref | PubMed | ISI | Google Scholar
  • 44. Christoffersen BO, Grand N, Golozoubova V, Svendsen O, Raun K. Gender-associated differences in metabolic syndrome-related parameters in Göttingen minipigs. Comp Med 57: 493–504, 2007.
    PubMed | ISI | Google Scholar
  • 45. Johansen T, Hansen HS, Richelsen B, Malmlöf R. The obese Göttingen minipig as a model of the metabolic syndrome: dietary effects on obesity, insulin sensitivity, and growth hormone profile. Comp Med 51: 150–155, 2001.
    PubMed | ISI | Google Scholar
  • 46. Ludvigsen TP, Kirk RK, Christoffersen BØ, Pedersen HD, Martinussen T, Kildegaard J, Heegaard PM, Lykkesfeldt J, Olsen LH. Göttingen minipig model of diet-induced atherosclerosis: influence of mild streptozotocin-induced diabetes on lesion severity and markers of inflammation evaluated in obese, obese and diabetic, and lean control animals. J Transl Med 13: 312 , 2015. doi:10.1186/s12967-015-0670-2.
    Crossref | PubMed | ISI | Google Scholar
  • 47. Sharp TE 3rd, Scarborough AL, Li Z, Polhemus DJ, Hidalgo HA, Schumacher JD, Matsuura TR, Jenkins JS, Kelly DP, Goodchild TT, Lefer DJ. Novel Göttingen miniswine model of heart failure with preserved ejection fraction integrating multiple comorbidities. JACC Basic Transl Sci 6: 154–170, 2021. doi:10.1016/j.jacbts.2020.11.012.
    Crossref | PubMed | Google Scholar
  • 48. Heinonen I, Sorop O, van Dalen BM, Wüst RCI, van de Wouw J, de Beer VJ, Octavia Y, van Duin RWB, Hoogstrate Y, Blonden L, Alkio M, Anttila K, Stubbs A, van der Velden J, Merkus D, Duncker DJ. Cellular, mitochondrial and molecular alterations associate with early left ventricular diastolic dysfunction in a porcine model of diabetic metabolic derangement. Sci Rep 10: 13173 , 2020. doi:10.1038/s41598-020-68637-4.
    Crossref | PubMed | ISI | Google Scholar
  • 49. van den Dorpel MMP, Heinonen I, Snelder SM, Vos HJ, Sorop O, van Domburg RT, Merkus D, Duncker DJ, van Dalen BM. Early detection of left ventricular diastolic dysfunction using conventional and speckle tracking echocardiography in a large animal model of metabolic dysfunction. Int J Cardiovasc Imaging 34: 743–749, 2018. doi:10.1007/s10554-017-1287-8.
    Crossref | PubMed | ISI | Google Scholar
  • 50. Andreasen LJ, Krog S, Ludvigsen TP, Nielsen OL, Møller JE, Christoffersen BØ, Pedersen HD, Olsen LH. Dietary normalization from a fat, fructose and cholesterol-rich diet to chow limits the amount of myocardial collagen in a Göttingen minipig model of obesity. Nutr Metab (Lond) 15: 64 , 2018. doi:10.1186/s12986-018-0303-x.
    Crossref | PubMed | ISI | Google Scholar
  • 51. Pedersen R, Ingerslev H-C, Sturek M, Alloosh M, Cirera S, Christoffersen BØ, Moesgaard SG, Larsen N, Boye M. Characterisation of gut microbiota in Ossabaw and Göttingen minipigs as models of obesity and metabolic syndrome. PLoS One 8: e56612 , 2013. doi:10.1371/journal.pone.0056612.
    Crossref | PubMed | ISI | Google Scholar
  • 52. Ludvigsen TP, Pedersen SF, Vegge A, Ripa RS, Johannesen HH, Hansen AE, Löfgren J, Schumacher-Petersen C, Kirk RK, Pedersen HD, Christoffersen BØ, Ørbæk M, Forman JL, Klausen TL, Olsen LH, Kjaer A. 18 F-FDG PET/MR-imaging in a Göttingen minipig model of atherosclerosis: correlations with histology and quantitative gene expression. Atherosclerosis 285: 55–63, 2019. doi:10.1016/j.atherosclerosis.2019.04.209.
    Crossref | PubMed | ISI | Google Scholar
  • 53. Rodríguez RR, González-Bulnes A, Garcia-Contreras C, Elena Rodriguez-Rodriguez A, Astiz S, Vazquez-Gomez M, Luis Pesantez J, Isabel B, Salido-Ruiz E, González J, Donate Correa J, Luis-Lima S, Porrini E. The Iberian pig fed with high-fat diet: a model of renal disease in obesity and metabolic syndrome. Int J Obes (Lond) 44: 457–465, 2020. doi:10.1038/s41366-019-0434-9.
    Crossref | PubMed | ISI | Google Scholar
  • 54. Torres-Rovira L, Astiz S, Caro A, Lopez-Bote C, Ovilo C, Pallares P, Perez-Solana ML, Sanchez-Sanchez R, Gonzalez-Bulnes A. Diet-induced swine model with obesity/leptin resistance for the study of metabolic syndrome and type 2 diabetes. ScientificWorldJournal 2012: 510149 , 2012. doi:10.1100/2012/510149.
    Crossref | PubMed | Google Scholar
  • 55. Fernández-Fígares I, Lachica M, Nieto R, Rivera-Ferre MG, Aguilera JF. Serum profile of metabolites and hormones in obese (Iberian) and lean (Landrace) growing gilts fed balanced or lysine deficient diets. Livest Sci 110: 73–81, 2007. doi:10.1016/j.livsci.2006.10.002.
    Crossref | ISI | Google Scholar
  • 56. Hernandez GV, Smith VA, Melnyk M, Burd MA, Sprayberry KA, Edwards MS, Peterson DG, Bennet DC, Fanter RK, Columbus DA, Steibel JP, Glanz H, Immoos C, Rice MS, Santiago-Rodriguez TM, Blank J, VanderKelen JJ, Kitts CL, Piccolo BD, La Frano MR, Burrin DG, Maj M, Manjarin R. Dysregulated FXR-FGF19 signaling and choline metabolism are associated with gut dysbiosis and hyperplasia in a novel pig model of pediatric NASH. Am J Physiol Gastrointest Liver Physiol 318: G582–G609, 2020. doi:10.1152/ajpgi.00344.2019.
    Link | ISI | Google Scholar
  • 57. Schwarzl M, Hamdani N, Seiler S, Alogna A, Manninger M, Reilly S, Zirngast B, Kirsch A, Steendijk P, Verderber J, Zweiker D, Eller P, Höfler G, Schauer S, Eller K, Maechler H, Pieske BM, Linke WA, Casadei B, Post H. A porcine model of hypertensive cardiomyopathy: implications for heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 309: H1407–H1418, 2015. doi:10.1152/ajpheart.00542.2015.
    Link | ISI | Google Scholar
  • 58. O'Donovan AN, Herisson FM, Fouhy F, Ryan PM, Whelan D, Johnson CN, Cluzel G, Ross RP, Stanton C, Caplice NM. Gut microbiome of a porcine model of metabolic syndrome and HF-pEF. Am J Physiol Heart Circ Physiol 318: H590–H603, 2020. doi:10.1152/ajpheart.00512.2019.
    Link | ISI | Google Scholar
  • 59. Zhang N, Feng B, Ma X, Sun K, Xu G, Zhou Y. Dapagliflozin improves left ventricular remodeling and aorta sympathetic tone in a pig model of heart failure with preserved ejection fraction. Cardiovasc Diabetol 18: 107 , 2019. doi:10.1186/s12933-019-0914-1.
    Crossref | PubMed | ISI | Google Scholar
  • 60. Hsu MC, Wang ME, Jiang YF, Liu HC, Chen YC, Chiu CH. Long-term feeding of high-fat plus high-fructose diet induces isolated impaired glucose tolerance and skeletal muscle insulin resistance in miniature pigs. Diabetol Metab Syndr 9: 81 , 2017. doi:10.1186/s13098-017-0281-6.
    Crossref | PubMed | ISI | Google Scholar
  • 61. Li SJ, Liu CH, Chang CW, Chu HP, Chen KJ, Mersmann HJ, Ding ST, Chu CH, Chen CY. Development of a dietary-induced metabolic syndrome model using miniature pigs involvement of AMPK and SIRT1. Eur J Clin Invest 45: 70–80, 2015. doi:10.1111/eci.12370.
    Crossref | PubMed | ISI | Google Scholar
  • 62. Li SJ, Wu TW, Chien MJ, Mersmann HJ, Chen CY. Involvement of pericardial adipose tissue in cardiac fibrosis of dietary-induced obese minipigs – role of mitochondrial function. Biochim Biophys Acta Mol Cell Biol Lipids 1864: 957–965, 2019. doi:10.1016/j.bbalip.2019.03.004.
    Crossref | PubMed | ISI | Google Scholar
  • 63. Li SJ, Liu CH, Chu HP, Mersmann HJ, Ding ST, Chu CH, Wang CY, Chen CY. The high-fat diet induces myocardial fibrosis in the metabolically healthy obese minipigs – the role of ER stress and oxidative stress. Clin Nutr 36: 760–767, 2017. doi:10.1016/j.clnu.2016.06.002.
    Crossref | PubMed | ISI | Google Scholar
  • 64. Li SJ, Ding ST, Mersmann HJ, Chu CH, Hsu CD, Chen CY. A nutritional nonalcoholic steatohepatitis minipig model. J Nutr Biochem 28: 51–60, 2016. doi:10.1016/j.jnutbio.2015.09.029.
    Crossref | PubMed | ISI | Google Scholar
  • 65. Nakamura N, Hatano E, Iguchi K, Sato M, Kawaguchi H, Ohtsu I, Sakurai T, Aizawa N, Iijima H, Nishiguchi S, Tomono T, Okuda Y, Wada S, Seo S, Taura K, Uemoto S, Ikegawa M. Elevated levels of circulating ITIH4 are associated with hepatocellular carcinoma with nonalcoholic fatty liver disease: from pig model to human study. BMC Cancer 19: 621 , 2019. doi:10.1186/s12885-019-5825-8.
    Crossref | PubMed | ISI | Google Scholar
  • 66. Yoshii D, Nakagawa T, Komohara Y, Kawaguchi H, Yamada S, Tanimoto A. Phenotypic changes in macrophage activation in a model of nonalcoholic fatty liver disease using microminipigs. J Atheroscler Thromb 28: 844–851, 2021. doi:10.5551/jat.57703.
    Crossref | PubMed | ISI | Google Scholar
  • 67. Yamada S, Kawaguchi H, Yamada T, Guo X, Matsuo K, Hamada T, Miura N, Tasaki T, Tanimoto A. Cholic acid enhances visceral adiposity, atherosclerosis and nonalcoholic fatty liver disease in microminipigs. J Atheroscler Thromb 24: 1150–1166, 2017. doi:10.5551/jat.39909.
    Crossref | PubMed | ISI | Google Scholar
  • 68. Yuan F, Guo L, Park KK-H, Woollard JR, Taek ‐Geun K, Jiang K, Melkamu T, Zang B, Smith SL, Fahrenkrug SC, Kolodgie FD, Lerman A, Virmani R, Lerman LO, Carlson DF. Ossabaw pigs with a PCSK9 gain-of-function mutation develop accelerated coronary atherosclerotic lesions: a novel model for preclinical studies. J Am Heart Assoc 7: e006207 , 2018. doi:10.1161/JAHA.117.006207.
    Crossref | PubMed | ISI | Google Scholar
  • 69. Kreutz RP, Alloosh M, Mansour K, Neeb Z, Kreutz Y, Flockhart DA, Sturek M. Morbid obesity and metabolic syndrome in Ossabaw miniature swine are associated with increased platelet reactivity. Diabetes Metab Syndr Obes 4: 99–105, 2011. doi:10.2147/DMSO.S17105.
    Crossref | PubMed | Google Scholar
  • 70. Badin JK, Kole A, Stivers B, Progar V, Pareddy A, Alloosh M, Sturek M. Alloxan-induced diabetes exacerbates coronary atherosclerosis and calcification in Ossabaw miniature swine with metabolic syndrome. J Transl Med 16: 58 , 2018. doi:10.1186/s12967-018-1431-9.
    Crossref | PubMed | ISI | Google Scholar
  • 71. Badin JK, Bruning RS, Sturek M. Effect of metabolic syndrome and aging on Ca2+ dysfunction in coronary smooth muscle and coronary artery disease severity in Ossabaw miniature swine. Exp Gerontol 108: 247–255, 2018. doi:10.1016/j.exger.2018.04.024.
    Crossref | PubMed | ISI | Google Scholar
  • 72. Badin JK, Progar V, Pareddy A, Cagle J, Alloosh M, Sturek M. Effect of age on diabetogenicity of alloxan in Ossabaw miniature swine. Comp Med 69: 114–122, 2019. doi:10.30802/AALAS-CM-18-000037.
    Crossref | PubMed | ISI | Google Scholar
  • 73. Bell LN, Lee L, Saxena R, Bemis KG, Wang M, Theodorakis JL, Vuppalanchi R, Alloosh M, Sturek M, Chalasani N. Serum proteomic analysis of diet-induced steatohepatitis and metabolic syndrome in the Ossabaw miniature swine. Am J Physiol Gastrointest Liver Physiol 298: G746–G754, 2010. doi:10.1152/ajpgi.00485.2009.
    Link | ISI | Google Scholar
  • 74. Bradley JM, Islam KN, Polhemus DJ, Donnarumma E, Brewster LP, Tao Y-X, Goodchild TT, Lefer DJ. Sustained release nitrite therapy results in myocardial protection in a porcine model of metabolic syndrome with peripheral vascular disease. Am J Physiol Heart Circ Physiol 309: H305–H317, 2015. doi:10.1152/ajpheart.00163.2015.
    Link | ISI | Google Scholar
  • 75. Bratz IN, Dick GM, Tune JD, Edwards JM, Neeb ZP, Dincer UD, Sturek M. Impaired capsaicin-induced relaxation of coronary arteries in a porcine model of the metabolic syndrome. Am J Physiol Heart Circ Physiol 294: H2489–H2496, 2008.doi:10.1152/ajpheart.01191.2007.
    Link | ISI | Google Scholar
  • 76. Choy JS, Luo T, Huo Y, Wischgoll T, Schultz K, Teague SD, Sturek M, Kassab GS. Compensatory enlargement of Ossabaw miniature swine coronary arteries in diffuse atherosclerosis. Int J Cardiol Heart Vasc 6: 4–11, 2015. doi:10.1016/j.ijcha.2014.11.003.
    Crossref | PubMed | Google Scholar
  • 77. Dincer UD. Cardiac β-adrenoceptor expression is markedly depressed in Ossabaw swine model of cardiometabolic risk. Int J Gen Med 4: 493–499, 2011. doi:10.2147/IJGM.S18175.
    Crossref | PubMed | Google Scholar
  • 78. Elmadhun NY, Lassaletta AD, Chu LM, Sellke FW. Metformin alters the insulin signaling pathway in ischemic cardiac tissue in a swine model of metabolic syndrome. J Thorac Cardiovasc Surg 145: 258–265, 2013.doi:10.1016/j.jtcvs.2012.09.028.
    Crossref | PubMed | ISI | Google Scholar
  • 79. Fullenkamp AM, Bell LN, Robbins RD, Lee L, Saxena R, Alloosh M, Klaunig JE, Mirmira RG, Sturek M, Chalasani N. Effect of different obesogenic diets on pancreatic histology in Ossabaw miniature swine. Pancreas 40: 438–443, 2011. doi:10.1097/MPA.0b013e3182061583.
    Crossref | PubMed | ISI | Google Scholar
  • 80. Handa RK, Liu Z, Connors BA, Alloosh M, Basile DP, Tune JD, Sturek M, Evan AP, Lingeman JE. Effect of renal shock wave lithotripsy on the development of metabolic syndrome in a juvenile swine model: a pilot study. J Urol 193: 1409–1416, 2015. doi:10.1016/j.juro.2014.09.037.
    Crossref | PubMed | ISI | Google Scholar
  • 81. Handa RK, Evan AP, Connors BA, Johnson CD, Liu Z, Alloosh M, Sturek M, Evans-Molina C, Mandeville JA, Gnessin E, Lingeman JE. Shock wave lithotripsy targeting of the kidney and pancreas does not increase the severity of metabolic syndrome in a porcine model. J Urol 192: 1257–1265, 2014. doi:10.1016/j.juro.2014.03.035.
    Crossref | PubMed | ISI | Google Scholar
  • 82. Hu G, Oboukhova EA, Kumar S, Sturek M, Obukhov AG. Canonical transient receptor potential channels expression is elevated in a porcine model of metabolic syndrome. Mol Endocrinol 23: 689–699, 2009. doi:10.1210/me.2008-0350.
    Crossref | PubMed | Google Scholar
  • 83. Lee L, Alloosh M, Saxena R, Van Alstine W, Watkins BA, Klaunig JE, Sturek M, Chalasani N. Nutritional model of steatohepatitis and metabolic syndrome in the Ossabaw miniature swine. Hepatology 50: 56–67, 2009. doi:10.1002/hep.22904.
    Crossref | PubMed | ISI | Google Scholar
  • 84. Liang T, Alloosh M, Bell LN, Fullenkamp A, Saxena R, Van Alstine W, Bybee P, Werling K, Sturek M, Chalasani N, Masuoka HC. Liver injury and fibrosis induced by dietary challenge in the Ossabaw miniature swine. PLoS One 10: e0124173 , 2015. doi:10.1371/journal.pone.0124173.
    Crossref | PubMed | ISI | Google Scholar
  • 85. Lim RR, Grant DG, Olver TD, Padilla J, Czajkowski AM, Schnurbusch TR, Mohan RR, Hainsworth DP, Walters EM, Chaurasia SS. Young Ossabaw pigs fed a western diet exhibit early signs of diabetic retinopathy. Invest Ophthalmol Vis Sci 59: 2325–2338, 2018. doi:10.1167/iovs.17-23616.
    Crossref | PubMed | Google Scholar
  • 86. Mattern HM, Lloyd PG, Sturek M, Hardin CD. Gender and genetic differences in bladder smooth muscle PPAR mRNA in a porcine model of the metabolic syndrome. Mol Cell Biochem 302: 43–49, 2007. doi:10.1007/s11010-007-9423-8.
    Crossref | PubMed | ISI | Google Scholar
  • 87. Newell-Fugate AE, Taibl JN, Alloosh M, Sturek M, Bahr JM, Nowak RA, Krisher RL. Effects of obesity and metabolic syndrome on steroidogenesis and folliculogenesis in the female Ossabaw mini-pig. PLoS One 10: e0128749 , 2015. doi:10.1371/journal.pone.0128749.
    Crossref | PubMed | ISI | Google Scholar
  • 88. Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, Emter CA. Western diet-fed, aortic-banded Ossabaw swine: a preclinical model of cardio-metabolic heart failure. JACC Basic Transl Sci 4: 404–421, 2019. doi:10.1016/j.jacbts.2019.02.004.
    Crossref | PubMed | Google Scholar
  • 89. Sabe AA, Elmadhun NY, Sadek AA, Chu LM, Bianchi C, Sellke FW. Differential effects of atorvastatin on autophagy in ischemic and nonischemic myocardium in Ossabaw swine with metabolic syndrome. J Thorac Cardiovasc Surg 148: 3172–3178, 2014. doi:10.1016/j.jtcvs.2014.07.104.
    Crossref | PubMed | ISI | Google Scholar
  • 90. Neeb ZP, Edwards JM, Alloosh M, Long X, Mokelke EA, Sturek M. Metabolic syndrome and coronary artery disease in Ossabaw compared with Yucatan swine. Comp Med 60: 300–315, 2010.
    PubMed | ISI | Google Scholar
  • 91. Westover AJ, Johnston KA, Buffington DA, Humes HD, David Humes H, Humes HD. An immunomodulatory device improves insulin resistance in obese porcine model of metabolic syndrome. J Diabetes Res 2016: 3486727 , 2016. doi:10.1155/2016/3486727.
    Crossref | PubMed | ISI | Google Scholar
  • 92. Hausman GJ, Martin RJ. Subcutaneous adipose tissue development in Yorkshire (lean) and Ossabaw (obese) pigs. J Anim Sci 52: 1442–1449, 1981. doi:10.2527/jas1981.5261442x.
    Crossref | PubMed | ISI | Google Scholar
  • 93. Lloyd PG, Sheehy AJ, Edwards JM, Mokelke EA, Sturek M. Leukemia inhibitory factor is upregulated in coronary arteries of Ossabaw miniature swine after stent placement. Coron Artery Dis 19: 217–226, 2008. doi:10.1097/MCA.0b013e3282f9d3be.
    Crossref | PubMed | ISI | Google Scholar
  • 94. Edwards JM, Neeb ZP, Alloosh MA, Long X, Bratz IN, Peller CR, Byrd JP, Kumar S, Obukhov AG, Sturek M. Exercise training decreases store-operated Ca2+ entry associated with metabolic syndrome and coronary atherosclerosis. Cardiovasc Res 85: 631–640, 2010. doi:10.1093/cvr/cvp308.
    Crossref | PubMed | ISI | Google Scholar
  • 95. Walker ME, Matthan NR, Solano-Aguilar G, Jang S, Lakshman S, Molokin A, Faits T, Urban JF, Johnson WE, Lamon-Fava S, Lichtenstein AH. A Western-type dietary pattern and atorvastatin induce epicardial adipose tissue interferon signaling in the Ossabaw pig. J Nutr Biochem 67: 212–218, 2019. doi:10.1016/J.JNUTBIO.2019.02.003.
    Crossref | PubMed | ISI | Google Scholar
  • 96. Uceda DE, Zhu X-Y, Woollard JR, Ferguson CM, Patras I, Carlson DF, Asirvatham SJ, Lerman A, Lerman LO. Accumulation of pericardial fat is associated with alterations in heart rate variability patterns in hypercholesterolemic pigs. Circ Arrhythm Electrophysiol 13: e007614 , 2020. doi:10.1161/CIRCEP.119.007614.
    Crossref | PubMed | ISI | Google Scholar
  • 97. Panasevich MR, Meers GM, Linden MA, Booth FW, Perfield JW, Fritsche KL, Wankhade UD, Chintapalli SV, Shankar K, Ibdah JA, Rector RS. High-fat, high-fructose, high-cholesterol feeding causes severe NASH and cecal microbiota dysbiosis in juvenile Ossabaw swine. Am J Physiol Endocrinol Physiol 314: E78–E92, 2018.doi:10.1152/ajpendo.00015.2017.
    Link | ISI | Google Scholar
  • 98. Ye S, Matthan NR, Lamon-Fava S, Aguilar GS, Turner JR, Walker ME, Chai Z, Lakshman S, Urban JF Jr, Lichtenstein AH. Western and heart healthy dietary patterns differentially affect the expression of genes associated with lipid metabolism, interferon signaling and inflammation in the jejunum of Ossabaw pigs. J Nutr Biochem 90: 108577 , 2021. doi:10.1016/j.jnutbio.2020.108577.
    Crossref | PubMed | ISI | Google Scholar
  • 99. Matthan NR, Solano-Aguilar G, Meng H, Lamon-Fava S, Goldbaum A, Walker ME, Jang S, Lakshman S, Molokin A, Xie Y, Beshah E, Stanley J, Urban JF, Lichtenstein AH. The Ossabaw pig is a suitable translational model to evaluate dietary patterns and coronary artery disease risk. J Nutr 148: 542–551, 2018. doi:10.1093/jn/nxy002.
    Crossref | PubMed | ISI | Google Scholar
  • 100. Vieira-Potter VJ, Lee S, Bayless DS, Scroggins RJ, Welly RJ, Fleming NJ, Smith TN, Meers GM, Hill MA, Scott Rector R, Padilla J. Disconnect between adipose tissue inflammation and cardiometabolic dysfunction in Ossabaw pigs. Obesity (Silver Spring) 23: 2421–2429, 2015. doi:10.1002/oby.21252.
    Crossref | PubMed | ISI | Google Scholar
  • 101. Faris RJ, Boddicker RL, Walker-Daniels J, Li J, Jones DE, Spurlock ME. Inflammation in response to n3 fatty acids in a porcine obesity model. Comp Med 62: 495–503, 2012.
    PubMed | ISI | Google Scholar
  • 102. Panasevich MR, Wankhade UD, Chintapalli SV, Shankar K, Rector RS. Cecal versus fecal microbiota in Ossabaw swine and implications for obesity. Physiol Genomics 50: 355–368, 2018. doi:10.1152/physiolgenomics.00110.2017.
    Link | ISI | Google Scholar
  • 103. Elmadhun NY, Sabe AA, Lassaletta AD, Chu LM, Kondra K, Sturek M, Sellke FW. Metabolic syndrome impairs notch signaling and promotes apoptosis in chronically ischemic myocardium. J Thorac Cardiovasc Surg 148: 1048–1055, 2014. doi:10.1016/j.jtcvs.2014.05.056.
    Crossref | PubMed | ISI | Google Scholar
  • 104. Li ZL, Woollard JR, Ebrahimi B, Crane JA, Jordan KL, Lerman A, Wang S-MM, Lerman LO. Progression from obesity to metabolic syndrome is associated with altered myocardial autophagy and apoptosis. Bone 23: 1–7, 2012. doi:10.1161/ATVBAHA.111.244061.
    Crossref | Google Scholar
  • 105. Borbouse L, Dick GM, Payne GA, Payne BD, Svendsen MC, Neeb ZP, Alloosh M, Bratz IN, Sturek M, Tune JD. Contribution of BKCa channels to local metabolic coronary vasodilation: effects of metabolic syndrome. Am J Physiol Heart Circ Physiol 298: H966–H973, 2010. doi:10.1152/ajpheart.00876.2009.
    Link | ISI | Google Scholar
  • 106. Berwick ZC, Dick GM, O’Leary HA, Bender SB, Goodwill AG, Moberly SP, Owen MK, Miller SJ, Obukhov AG, Tune JD. Contribution of electromechanical coupling between Kv and Ca v1.2 channels to coronary dysfunction in obesity. Basic Res Cardiol 108: 370 , 2013. doi:10.1007/s00395-013-0370-0.
    Crossref | PubMed | ISI | Google Scholar
  • 107. Moberly SP, Mather KJ, Berwick ZC, Owen MK, Goodwill AG, Casalini ED, Hutchins GD, Green MA, Ng Y, Considine RV, Perry KM, Chisholm RL, Tune JD. Impaired cardiometabolic responses to glucagon-like peptide 1 in obesity and type 2 diabetes mellitus. Basic Res Cardiol 108: 365 , 2013. doi:10.1007/s00395-013-0365-x.
    Crossref | PubMed | ISI | Google Scholar
  • 108. Berwick ZC, Dick GM, Moberly SP, Kohr MC, Sturek M, Tune JD. Contribution of voltage-dependent K+ channels to metabolic control of coronary blood flow. J Mol Cell Cardiol 52: 912–919, 2012. doi:10.1016/j.yjmcc.2011.07.004.
    Crossref | PubMed | ISI | Google Scholar
  • 109. Kelly SC, Rau CD, Ouyang A, Thorne PK, Olver TD, Edwards JC, Domeier TL, Padilla J, Grisanti LA, Fleenor BS, Wang Y, Rector RS, Emter CA. The right ventricular transcriptome signature in Ossabaw swine with cardiometabolic heart failure: implications for the coronary vasculature. Physiol Genomics 53: 99–115, 2021. doi:10.1152/physiolgenomics.00093.2020.
    Link | ISI | Google Scholar
  • 110. Lassaletta AD, Chu LM, Robich MP, Elmadhun NY, Feng J, Burgess TA, Laham RJ, Sturek M, Sellke FW. Overfed Ossabaw swine with early stage metabolic syndrome have normal coronary collateral development in response to chronic ischemia. Basic Res Cardiol 107: 243 , 2012. doi:10.1007/s00395-012-0243-y.
    Crossref | PubMed | ISI | Google Scholar
  • 111. Dyson MC, Alloosh M, Vuchetich JP, Mokelke EA, Sturek M. Components of metabolic syndrome and coronary artery disease in female Ossabaw swine fed excess atherogenic diet. Comp Med 56: 35–45, 2006.
    PubMed | ISI | Google Scholar
  • 112. Wastney M, Lee W, Jackson GS, Alloosh M, Sturek M, Lachcik P, Peacock M, Martin B, Weaver CM. Soft tissue calcification in the Ossabaw miniature pig: experimental and kinetic modeling studies. Osteoporos Int 24: 2123–2126, 2013. doi:10.1007/s00198-012-2229-x.
    Crossref | PubMed | ISI | Google Scholar
  • 113. Burgess TA, Robich MP, Chu LM, Bianchi C, Sellke FW. Improving glucose metabolism with resveratrol in a swine model of metabolic syndrome through alteration of signaling pathways in the liver and skeletal muscle. Arch Surg 146: 556–564, 2011. doi:10.1001/archsurg.2011.100.
    Crossref | PubMed | Google Scholar
  • 114. Elmadhun NY, Lassaletta AD, Chu LM, Bianchi C, Sellke FW. Vodka and wine consumption in a swine model of metabolic syndrome alters insulin signaling pathways in the liver and skeletal muscle. Surg (United States) 152: 414–422, 2012. doi:10.1016/j.surg.2012.06.014.
    Crossref | PubMed | ISI | Google Scholar
  • 115. Karimi M, Pavlov VI, Ziegler O, Sriram N, Yoon SY, Agbortoko V, Alexandrova S, Asara J, Sellke FW, Sturek M, Feng J, Alexandrov BS, Usheva A. Robust effect of metabolic syndrome on major metabolic pathways in the myocardium. PLoS One 14: e0225857 , 2019. doi:10.1371/journal.pone.0225857.
    Crossref | PubMed | ISI | Google Scholar
  • 116. Liao S, Zhou Q, Zhang Y. Elastic aortic wrap reduced aortic stiffness by partially alleviating the impairment of cholesterol efflux capacity in pigs. J Diabetes Metab Disord 17: 101–109, 2018. doi:10.1007/s40200-018-0345-7.
    Crossref | PubMed | ISI | Google Scholar
  • 117. Matyal R, Chu L, Mahmood F, Robich MP, Wang A, Hess PE, Shahul S, Pinto DS, Khabbaz K, Sellke FW. Neuropeptide Y improves myocardial perfusion and function in a swine model of hypercholesterolemia and chronic myocardial ischemia. J Mol Cell Cardiol 53: 891–898, 2012. doi:10.1016/j.yjmcc.2012.08.027.
    Crossref | PubMed | ISI | Google Scholar
  • 118. Robich MP, Chu LM, Burgess TA, Feng J, Han Y, Nezafat R, Leber MP, Laham RJ, Manning WJ, Sellke FW. Resveratrol preserves myocardial function and perfusion in remote nonischemic myocardium in a swine model of metabolic syndrome. J Am Coll Surg 215: 681–689, 2012. doi:10.1016/j.jamcollsurg.2012.06.417.
    Crossref | PubMed | ISI | Google Scholar
  • 119. Robich MP, Chu LM, Chaudray M, Nezafat R, Han Y, Clements RT, Laham RJ, Manning WJ, Coady MA, Sellke FW. Anti-angiogenic effect of high-dose resveratrol in a swine model of metabolic syndrome. Surgery 148: 453–462, 2010. doi:10.1016/j.surg.2010.04.013.
    Crossref | PubMed | ISI | Google Scholar
  • 120. Robich MP, Osipov RM, Chu LM, Han Y, Feng J, Nezafat R, Clements RT, Manning WJ, Sellke FW. Resveratrol modifies risk factors for coronary artery disease in swine with metabolic syndrome and myocardial ischemia. Eur J Pharmacol 664: 45–53, 2011. doi:10.1016/j.ejphar.2011.04.059.
    Crossref | PubMed | ISI | Google Scholar
  • 121. Sabe AA, Sadek AA, Elmadhun NY, Dalal RS, Robich MP, Bianchi C, Sellke FW. Investigating the effects of resveratrol on chronically ischemic myocardium in a swine model of metabolic syndrome: a proteomics analysis. J Med Food 18: 60–66, 2015. doi:10.1089/jmf.2014.0036.
    Crossref | PubMed | ISI | Google Scholar
  • 122. Saraf R, Huang T, Mahmood F, Owais K, Bardia A, Khabbaz KR, Liu D, Senthilnathan V, Lassaletta AD, Sellke F, Matyal R. Early cellular changes in the ascending aorta and myocardium in a swine model of metabolic syndrome. PLoS One 11: e0146481 , 2016. doi:10.1371/journal.pone.0146481.
    Crossref | PubMed | ISI | Google Scholar
  • 123. Lawandy I, Liu Y, Shi G, Zhang Z, Scrimgeour LA, Pavlov V, Jaworski R, Sellke FW, Feng J, Pavlov V, Jaworski R, Sellke FW, Feng J, Pavlov V, Jaworski R, Sellke FW, Feng J. Increased coronary arteriolar contraction to serotonin in juvenile pigs with metabolic syndrome. Mol Cell Biochem 461: 57–64, 2019. PMC]doi:10.1007/s11010-019-03589-6.
    Crossref | PubMed | ISI | Google Scholar
  • 124. Te Pas MFW, Koopmans SJ, Kruijt L, Calus MPL, Smits MA. Plasma proteome profiles associated with diet-induced metabolic syndrome and the early onset of metabolic syndrome in a pig model. PLoS One 8: e73087 , 2013. doi:10.1371/journal.pone.0073087.
    Crossref | PubMed | ISI | Google Scholar
  • 125. van den Heuvel M, Sorop O, Koopmans SJ, Dekker R, de Vries R, van Beusekom HM, Eringa EC, Duncker DJ, Danser AH, van der Giessen WJ. Coronary microvascular dysfunction in a porcine model of early atherosclerosis and diabetes. Am J Physiol Heart Circ Physiol 302: H85–H94, 2012. doi:10.1152/ajpheart.00311.2011.
    Link | ISI | Google Scholar
  • 126. Sorop O, Heinonen I, van Kranenburg M, van de Wouw J, de Beer VJ, Nguyen ITN, Octavia Y, van Duin RWB, Stam K, van Geuns RJ, Wielopolski PA, Krestin GP, van den Meiracker AH, Verjans R, van Bilsen M, Danser AHJ, Paulus WJ, Cheng C, Linke WA, Joles JA, Verhaar MC, van der Velden J, Merkus D, Duncker DJ. Multiple common comorbidities produce left ventricular diastolic dysfunction associated with coronary microvascular dysfunction, oxidative stress, and myocardial stiffening. Cardiovasc Res 114: 954–964, 2018. doi:10.1093/cvr/cvy038.
    Crossref | PubMed | ISI | Google Scholar
  • 127. Pedersen R, Andersen AD, Hermann-Bank ML, Stagsted J, Boye M. Gut Microbes The effect of high-fat diet on the composition of the gut microbiota in cloned and non-cloned pigs of lean and obese phenotype. Gut Microbes 4: 371–381, 2013. doi:10.4161/gmic.26108.
    Crossref | PubMed | ISI | Google Scholar
  • 128. Curtasu MV, Knudsen KEB, Callesen H, Purup S, Stagsted J, Hedemann MS. Obesity development in a miniature Yucatan pig model: a multi-compartmental metabolomics study on cloned and normal pigs fed restricted or ad libitum high-energy diets. J Proteome Res 18: 30–47, 2019. doi:10.1021/acs.jproteome.8b00264.
    Crossref | PubMed | ISI | Google Scholar
  • 129. Huang JV, Lu L, Ye S, Bergman BC, Sparagna GC, Sarraf M, Reusch JEB, Greyson CR, Schwartz GG. Impaired contractile recovery after low-flow myocardial ischemia in a porcine model of metabolic syndrome. Am J Physiol Heart Circ Physiol 304: H861–H873, 2013. doi:10.1152/ajpheart.00535.2012.
    Link | ISI | Google Scholar
  • 130. Lee J, Xu Y, Lu L, Bergman B, Leitner JW, Greyson C, Draznin B, Schwartz GG. Multiple abnormalities of myocardial insulin signaling in a porcine model of diet-induced obesity. Am J Physiol Heart Circ Physiol 298: H310–H319, 2010. doi:10.1152/ajpheart.00359.2009.
    Link | ISI | Google Scholar
  • 131. Pop R, Kong SH, Langlois A, Marchegiani F, Shlomovitz E, Legnèr A, Bietiger W, Pinget M, Beaujeux R, Mutter D, Marescaux J, Diana M. Gastrointestinal hormones manipulation to counteract metabolic syndrome using duodenal targeted embolization. Surg Innov 26: 280–292, 2019. doi:10.1177/1553350619838098.
    Crossref | PubMed | ISI | Google Scholar
  • 132. Marshall KD, Muller BN, Krenz M, Hanft LM, McDonald KS, Dellsperger KC, Emter CA. Heart failure with preserved ejection fraction: chronic low-intensity interval exercise training preserves myocardial O2 balance and diastolic function. J Appl Physiol 114: 131–147, 2013. doi:10.1152/japplphysiol.01059.2012.
    Link | ISI | Google Scholar
  • 133. Yongming P, Zhaowei C, Yichao M, Keyan Z, Liang C, Fangming C, Xiaoping X, Quanxin M, Minli C. Involvement of peroxisome proliferator-activated receptors in cardiac and vascular remodeling in a novel minipig model of insulin resistance and atherosclerosis induced by consumption of a high-fat/cholesterol diet. Cardiovasc Diabetol 14: 6 , 2015. doi:10.1186/s12933-014-0165-0.
    Crossref | PubMed | ISI | Google Scholar
  • 134. Swindle MM, Makin A, Herron AJ, Clubb FJ, Frazier KS. Swine as models in biomedical research and toxicology testing. Vet Pathol 49: 344–356, 2012 [Erratum in Vet Pathol 49: 738, 2012]. doi:10.1177/0300985811402846.
    Crossref | PubMed | ISI | Google Scholar
  • 135. Sipos W. Shifts in porcine PBMC populations from adolescence to adulthood. Vet Immunol Immunopathol 211: 35–37, 2019. doi:10.1016/j.vetimm.2019.04.002.
    Crossref | PubMed | ISI | Google Scholar
  • 136. Barbero A, Astiz S, Lopez-Bote CJ, Perez-Solana ML, Ayuso M, Garcia-Real I, Gonzalez-Bulnes A. Maternal malnutrition and offspring sex determine juvenile obesity and metabolic disorders in a swine model of leptin resistance. PLoS One 8: e78424 , 2013. doi:10.1371/journal.pone.0078424.
    Crossref | PubMed | ISI | Google Scholar
  • 137. Taylor G, Roese G, Hermesch S. Breeds of pigs-Landrace Origin and history Breed characteristics (Online). www.dpi.nsw.gov.au [ 2021 Jun 15 ].
    Google Scholar
  • 138. Briggs HM. International Pig Breed Encyclopedia (Online). https://books.google.ie/books/about/International_Pig_Breed_Encyclopedia.html?id=kcEctwAACAAJ&redir_esc=y [ 2021 Jun 15 ].
    Google Scholar
  • 139. Gerrity RG, Natarajan R, Nadler JL, Kimsey T. Diabetes-induced accelerated atherosclerosis in swine. Diabetes 50: 1654–1665, 2001. doi:10.2337/diabetes.50.7.1654.
    Crossref | PubMed | ISI | Google Scholar
  • 140. Sturek M, Alloosh M, Wenzel J, Byrd J, Edwards J, Lloyd P, Tune J, March K, Miller M, Mokelke E, Lehr Brisbin I Jr. Ossabaw island miniature swine: cardiometabolic syndrome assessment. In: Swine in the Laboratory: Surgery, Anesthesia, Imaging, and Experimental Techniques. Boca Raton, FL: CRC Press, 2007, p. 397–402.
    Crossref | Google Scholar
  • 141. Sturek M, Tune JD, Alloosh M. Ossabaw Island miniature swine: metabolic syndrome and cardiovascular assessment. In: Swine in the Laboratory: Surgery, Anesthesia, Imaging, and Experimental Techniques, edited by Swindle MM, Smith AC. Boca Raton, FL: CRC Press, 2015, p. 451–465.
    Crossref | Google Scholar
  • 142. Zhang Y, Fan G, Liu X, Skovgaard K, Sturek M, Heegaard PMH. The genome of the naturally evolved obesity-prone Ossabaw miniature pig. iScience 24: 103081 , 2021. doi:10.1016/j.isci.2021.103081.
    Crossref | PubMed | ISI | Google Scholar
  • 143. Lee TY, Sung YY, Huang TM. New Miniature pig Lee-Sung Strain in Taiwan (Online). https://agris.fao.org/agris-search/search.do?recordID=US201301396794 [ 2021 May 21 ].
    Google Scholar
  • 144. Jiang YN, Wu CY, Huang CY, Chu HP, Ke MW, Kung MS, Li KY, Wang CH, Li SH, Wang Y, Ju YT. Interpopulation and intrapopulation maternal lineage genetics of the Lanyu pig (Sus scrofa) by analysis of mitochondrial cytochrome b and control region sequences. J Anim Sci 86: 2461–2470, 2008. doi:10.2527/jas.2007-0049.
    Crossref | PubMed | ISI | Google Scholar
  • 145. Liu Y, Zeng BH, Shang HT, Cen YY, Wei H. Bama miniature pigs (sus scrofa domestica) as a model for drug evaluation for humans: comparison of in vitro metabolism and in vivo pharmacokinetics of lovastatin. Comp Med 58: 580–587, 2008.
    PubMed | ISI | Google Scholar
  • 146. Gao C, Jiang Q, Guo D, Liu J, Han L, Qu L. Characterization of swine leukocyte antigen (SLA) polymorphism by sequence-based and PCR-SSP methods in Chinese Bama miniature pigs. Dev Comp Immunol 45: 87–96, 2014. doi:10.1016/j.dci.2014.02.006.
    Crossref | PubMed | ISI | Google Scholar
  • 147. Li X, Mao M, Zhang Y, Yu K, Zhu W. Succinate modulates intestinal barrier function and inflammation response in pigs. Biomolecules 9: 486 , 2019. doi:10.3390/biom9090486.
    Crossref | PubMed | ISI | Google Scholar
  • 148. Chen H, Liu YQ, Li CH, Guo XM, Huang LJ. The susceptibility of three strains of Chinese minipigs to diet-induced type 2 diabetes mellitus. Lab Anim (NY) 38: 355–363, 2009. doi:10.1038/laban1109-355.
    Crossref | PubMed | ISI | Google Scholar
  • 149. Panepinto LM, Phillips RW, Wheeler LR, Will. DH. The Yucatan minature pig as a laboratory animal. Lab Anim Sci 28: 308–313, 1978. doi:10.2527/jas.2007-0220.
    Crossref | PubMed | Google Scholar
  • 150. Bekiares NA, Chen AS, Shanmuganayagam D, Meyers AD, Crenshaw TD, Krueger CG, Reed JD. Effect of caloric restriction on metabolic dysfunction of young rapacz familial hypercholesterolemic swine (Sus scrofa). Comp Med 67: 508–517, 2017.
    PubMed | ISI | Google Scholar
  • 151. Iversen MW, Nordbø Ø, Gjerlaug-Enger E, Grindflek E, Lopes MS, Meuwissen T. Effects of heterozygosity on performance of purebred and crossbred pigs. Genet Sel Evol 51: 8 , 2019. doi:10.1186/s12711-019-0450-1.
    Crossref | PubMed | ISI | Google Scholar
  • 152. Roth JA, Tuggle CK. Livestock models in translational medicine. ILAR J 56: 1–6, 2015. doi:10.1093/ilar/ilv011.
    Crossref | PubMed | ISI | Google Scholar
  • 153. Gerstein HC, Waltman L. Why don’t pigs get diabetes? Explanations for variations in diabetes susceptibility in human populations living in a diabetogenic environment. CMAJ 174: 25–26, 2006. doi:10.1503/cmaj.050649.
    Crossref | PubMed | ISI | Google Scholar
  • 154. Renner S, Blutke A, Clauss S, Deeg CA, Kemter E, Merkus D, Wanke R, Wolf E. Porcine models for studying complications and organ crosstalk in diabetes mellitus. Cell Tissue Res 380: 341–378, 2020. doi:10.1007/s00441-019-03158-9.
    Crossref | PubMed | ISI | Google Scholar
  • 155. King AJF. The use of animal models in diabetes research. Br J Pharmacol 166: 877–894, 2012. doi:10.1111/j.1476-5381.2012.01911.x.
    Crossref | PubMed | ISI | Google Scholar
  • 156. Laguens RP, Candela S, Hernández RE, Gagliardino JJ. Streptozotocin-induced liver damage in mice. Horm Metab Res 12: 197–201, 1980. doi:10.1055/s-2007-996241.
    Crossref | PubMed | ISI | Google Scholar
  • 157. Wold LE, Ren J. Streptozotocin directly impairs cardiac contractile function in isolated ventricular myocytes via a p38 map kinase-dependent oxidative stress mechanism. Biochem Biophys Res Commun 318: 1066–1071, 2004. doi:10.1016/j.bbrc.2004.04.138.
    Crossref | PubMed | ISI | Google Scholar
  • 158. Evan AP, Mong SA, Gattone VH, Connors BA, Aronoff GR, Luft FC. The effect of streptozotocin and streptozotocin-induced diabetes on the kidney. Ren Physiol 7: 78–89, 1984. doi:10.1159/000172927.
    Crossref | PubMed | Google Scholar
  • 159. Dufrane D, van Steenberghe M, Guiot Y, Goebbels RM, Saliez A, Gianello P. Streptozotocin-induced diabetes in large animals (pigs/primates): role of GLUT2 transporter and β-cell plasticity. Transplantation 81: 36–45, 2006. doi:10.1097/01.tp.0000189712.74495.82.
    Crossref | PubMed | ISI | Google Scholar
  • 160. Hara H, Lin YJ, Zhu X, Tai H-C, Ezzelarab M, Balamurugan AN, Balamarugan AN, Bottino R, Houser SL, Cooper DKC. Safe induction of diabetes by high-dose streptozotocin in pigs. Pancreas 36: 31–38, 2008 [Erratum in Pancreas 36: 328, 2008]. doi:10.1097/mpa.0b013e3181452886.
    Crossref | PubMed | ISI | Google Scholar
  • 161. Furman BL. Streptozotocin‐induced diabetic models in mice and rats. Curr Protoc 1: e78 , 2021. doi:10.1002/cpz1.78.
    Crossref | PubMed | Google Scholar
  • 162. Tomek J, Bub G. Hypertension-induced remodelling: on the interactions of cardiac risk factors. J Physiol 595: 4027–4036, 2017. doi:10.1113/JP273043.
    Crossref | PubMed | ISI | Google Scholar
  • 163. Kumar P, Gehi AK. Atrial fibrillation and metabolic syndrome: understanding the connection. J Atr Fibrillation 5: 647 , 2012. doi:10.4022/jafib.647.
    Crossref | PubMed | Google Scholar
  • 164. Chakaroun RM, Massier L, Kovacs P. Gut microbiome, intestinal permeability, and tissue bacteria in metabolic disease: perpetrators or bystanders? Nutrients 12: 1082 , 2020. doi:10.3390/nu12041082.
    Crossref | PubMed | ISI | Google Scholar
  • 165. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BAH, Forslund K, Hildebrand F, Prifti E, Falony G, Le Chatelier E, Levenez F, Doré J, Mattila I, Plichta DR, Pöhö P, Hellgren LI, Arumugam M, Sunagawa S, Vieira-Silva S, Jørgensen T, Holm JB, Trošt K; MetaHIT Consortium , Kristiansen K, Brix S, Raes J, Wang J, Hansen T, Bork P, Brunak S, Oresic M, Ehrlich SD, Pedersen O. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535: 376–381, 2016. doi:10.1038/nature18646.
    Crossref | PubMed | ISI | Google Scholar
  • 166. van der Beek CM, Canfora EE, Kip AM, Gorissen SHM, Olde Damink SWM, van Eijk HM, Holst JJ, Blaak EE, Dejong CHC, Lenaerts K. The prebiotic inulin improves substrate metabolism and promotes short-chain fatty acid production in overweight to obese men. Metabolism 87: 25–35, 2018. doi:10.1016/j.metabol.2018.06.009.
    Crossref | PubMed | ISI | Google Scholar
  • 167. Ryan PM, Stanton C, Caplice NM. Bile acids at the cross-roads of gut microbiome-host cardiometabolic interactions. Diabetol Metab Syndr 9: 1–12, 2017. doi:10.1186/s13098-017-0299-9.
    Crossref | PubMed | ISI | Google Scholar
  • 168. Li HH, Li YP, Zhu Q, Qiao JY, Wang WJ. Dietary supplementation with Clostridium butyricum helps to improve the intestinal barrier function of weaned piglets challenged with enterotoxigenic Escherichia coli K88. J Appl Microbiol 125: 964–975, 2018. doi:10.1111/jam.13936.
    Crossref | PubMed | ISI | Google Scholar
  • 169. de Diego-Cabero N, Mereu A, Menoyo D, Holst JJ, Ipharraguerre IR. Bile acid mediated effects on gut integrity and performance of early-weaned piglets. BMC Vet Res 11: 111 , 2015. doi:10.1186/s12917-015-0425-6.
    Crossref | PubMed | ISI | Google Scholar
  • 170. Myhill LJ, Stolzenbach S, Hansen TVA, Skovgaard K, Stensvold CR, Andersen LOB, Nejsum P, Mejer H, Thamsborg SM, Williams AR. Mucosal barrier and Th2 immune responses are enhanced by dietary inulin in pigs infected with trichuris suis. Front Immunol 9: 2557 , 2018. doi:10.3389/fimmu.2018.02557.
    Crossref | PubMed | ISI | Google Scholar
  • 171. Castaner O, Goday A, Park YM, Lee SH, Magkos F, Shiow SATE, Schröder H. The gut microbiome profile in obesity: a systematic review. Int J Endocrinol 2018: 4095789 , 2018. doi:10.1155/2018/4095789.
    Crossref | PubMed | ISI | Google Scholar
  • 172. Xiao L, Estellé J, Kiilerich P, Ramayo-Caldas Y, Xia Z, Feng Q, Liang S, Pedersen A, Kjeldsen NJ, Liu C, Maguin E, Doré J, Pons N, Le Chatelier E, Prifti E, Li J, Jia H, Liu X, Xu X, Ehrlich SD, Madsen L, Kristiansen K, Rogel-Gaillard C, Wang J. A reference gene catalogue of the pig gut microbiome. Nat Microbiol 1: 1–6, 2016. doi:10.1038/nmicrobiol.2016.161.
    Crossref | PubMed | ISI | Google Scholar
  • 173. Guevarra RB, Kim J, Nguyen SG, Unno T. Comparison of fecal microbial communities between white and black pigs. JABC 58: 369–375, 2015. doi:10.3839/jabc.2015.058.
    Crossref | Google Scholar
  • 174. Fernandez-Real JM, Serino M, Blasco G, Puig J, Daunis-I-Estadella J, Ricart W, Burcelin R, Fernández-Aranda F, Portero-Otin M. Gut microbiota interacts with brain microstructure and function. J Clin Endocrinol Metab 100: 4505–4513, 2015. doi:10.1210/jc.2015-3076.
    Crossref | PubMed | ISI | Google Scholar
  • 175. Baranowski BJ, Allen MD, Nyarko JNK, Rector RS, Padilla J, Mousseau DD, Rau CD, Wang Y, Laughlin MH, Emter CA, MacPherson REK, Olver TD. Cerebrovascular insufficiency and amyloidogenic signaling in Ossabaw swine with cardiometabolic heart failure. JCI Insight 6: e143141 , 2021. doi:10.1172/jci.insight.143141.
    Crossref | PubMed | ISI | Google Scholar
  • 176. Olver TD, Grunewald ZI, Jurrissen TJ, MacPherson REK, Leblanc PJ, Schnurbusch TR, Czajkowski AM, Laughlin MH, Rector RS, Bender SB, Walters EM, Emter CA, Padilla J. Microvascular insulin resistance in skeletal muscle and brain occurs early in the development of juvenile obesity in pigs. Am J Physiol Regul Integr Comp Physiol 314: R252–R264, 2018. doi:10.1152/ajpregu.00213.2017.
    Link | ISI | Google Scholar

AUTHOR NOTES