1
|
Lin C, Dong Z, Song J, Wang S, Yang Y, Li H, Feng Z, Pei Y. Differences in histomorphology and expression of key lipid regulated genes of four adipose tissues from Tibetan pigs. PeerJ 2023; 11:e14556. [PMID: 36643642 PMCID: PMC9835692 DOI: 10.7717/peerj.14556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/21/2022] [Indexed: 01/11/2023] Open
Abstract
Tibetan pigs, an indigenous pig breed in China, have high overall fat deposition and flavorful and tasty meat. They are thus good models for studying adipogenesis. Few studies have been conducted focusing on expression of lipid regulated genes in different adipose tissues of Tibetan pigs. Therefore, we compared the difference of histomorphology and expression level of lipid regulated genes through qPCR and western blot in subcutaneous fat, perirenal fat, omental adipose tissue, and inguinal fat of Tibetan pigs. Our results showed that the area of subcutaneous adipocytes in Tibetan pigs was smaller, while the other three adipose tissues (perirenal fat, greater omentum fat, inguinal fat) had cell areas of similar size. The gene expression of FABP4, FASN, FABP3, and ME1 in subcutaneous fat was significantly higher than that in perirenal fat. Furthermore, the protein expression of FABP4 was significantly lower in subcutaneous fat than in perirenal fat (p < 0.05), and the expression of FASN was higher in greater omentum fat than in subcutaneous fat (p = 0.084). The difference in adipocyte cell size and expression of lipid-regulated genes in adipose tissues from the various parts of the pig body is likely due to the different cellular lipid metabolic processes. Specially, FABP4 and FASN may be involved in the regulation of fat deposition in different adipose tissues of Tibetan pigs.
Collapse
Affiliation(s)
- Chenghong Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zexia Dong
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Jia Song
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Sutian Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Ying Yang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Feng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yangli Pei
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
2
|
Moore MP, Cunningham RP, Meers GM, Johnson SA, Wheeler AA, Ganga RR, Spencer NM, Pitt JB, Diaz-Arias A, Swi AIA, Hammoud GM, Ibdah JA, Parks EJ, Rector RS. Compromised hepatic mitochondrial fatty acid oxidation and reduced markers of mitochondrial turnover in human NAFLD. Hepatology 2022; 76:1452-1465. [PMID: 35000203 PMCID: PMC9270503 DOI: 10.1002/hep.32324] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS NAFLD and its more-advanced form, steatohepatitis (NASH), is associated with obesity and is an independent risk factor for cardiovascular, liver-related, and all-cause mortality. Available human data examining hepatic mitochondrial fatty acid oxidation (FAO) and hepatic mitochondrial turnover in NAFLD and NASH are scant. APPROACH AND RESULTS To investigate this relationship, liver biopsies were obtained from patients with obesity undergoing bariatric surgery and data clustered into four groups based on hepatic histopathological classification: Control (CTRL; no disease); NAFL (steatosis only); Borderline-NASH (steatosis with lobular inflammation or hepatocellular ballooning); and Definite-NASH (D-NASH; steatosis, lobular inflammation, and hepatocellular ballooning). Hepatic mitochondrial complete FAO to CO2 and the rate-limiting enzyme in β-oxidation (β-hydroxyacyl-CoA dehydrogenase activity) were reduced by ~40%-50% with D-NASH compared with CTRL. This corresponded with increased hepatic mitochondrial reactive oxygen species production, as well as dramatic reductions in markers of mitochondrial biogenesis, autophagy, mitophagy, fission, and fusion in NAFL and NASH. CONCLUSIONS These findings suggest that compromised hepatic FAO and mitochondrial turnover are intimately linked to increasing NAFLD severity in patients with obesity.
Collapse
Affiliation(s)
- Mary P. Moore
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Rory P. Cunningham
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Grace M. Meers
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Sarah A. Johnson
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Andrew A. Wheeler
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - Rama R. Ganga
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - Nicole M. Spencer
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - James B. Pitt
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | | | - Ahmed I. A. Swi
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Ghassan M. Hammoud
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Jamal A. Ibdah
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Elizabeth J. Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - R. Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| |
Collapse
|
3
|
Salamun V, Rizzo M, Lovrecic L, Hocevar K, Papler Burnik T, Janez A, Jensterle M, Vrtacnik Bokal E, Peterlin B, Maver A. The Endometrial Transcriptome of Metabolic and Inflammatory Pathways During the Window of Implantation Is Deranged in Infertile Obese Polycystic Ovarian Syndrome Women. Metab Syndr Relat Disord 2022; 20:384-394. [PMID: 35834645 DOI: 10.1089/met.2021.0149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction and Aim: Obese women with polycystic ovarian syndrome (PCOS) have a reduced rate of spontaneous conception even when their cycles are ovulatory. Endometrial receptivity is an important factor for poor implantation and increased miscarriage rates. Mechanisms in which both pathologies modify the endometrium are not fully clarified. The aim of our study was to compare the endometrial transcriptomic profiles between infertile obese PCOS (O-PCOS) women and infertile normal weight subjects during the window of implantation in ovulatory menstrual cycles. Methods: We conducted a prospective transcriptomic analysis of the endometrium using RNA sequencing. In this way, potential endometrial mechanisms leading to the poor reproductive outcome in O-PCOS patients could be characterized. Endometrial samples during days 21-23 of the menstrual cycle were collected from infertile O-PCOS women (n = 11) and normal weight controls (n = 10). Subgroups were defined according to the ovulatory/anovulatory status in the natural cycles, and O-PCOS women were grouped into the O-PCOS ovulatory (O-PCOS-ovul) subgroup. RNA isolation, sequencing with library reparation, and subsequent RNAseq data analysis were performed. Results: Infertile O-PCOS patients had 610 differentially expressed genes (DEGs), after adjustment for multiple comparisons with normal weight infertile controls, related to obesity (MXRA5 and ECM1), PCOS (ADAMTS19 and SLC18A2), and metabolism (VNN1 and PC). In the ovulatory subgroup, no DEGs were found, but significant differences in canonical pathways and the upstream regulator were revealed. According to functional and upstream analyses of ovulatory subgroup comparisons, the most important biological processes were related to inflammation (TNFR1 signaling), insulin signaling (insulin receptor signaling and PI3/AKT), fatty acid metabolism (stearate biosynthesis I and palmitate biosynthesis I), and lipotoxicity (unfolded protein response pathway). Conclusions: We demonstrated that endometrial transcription in ovulatory O-PCOS patients is deranged in comparison with the control ovulatory endometrium. The most important pathways of differentiation include metabolism and inflammation. These processes could also represent potential mechanisms for poor embryo implantation, which prevent the development of a successful pregnancy. ClinicalTrials.gov ID: NCT03353948.
Collapse
Affiliation(s)
- Vesna Salamun
- Division of Obstetrics and Gynecology, Department of Human Reproduction, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Manfredi Rizzo
- Division of Endocrinology, Diabetes, and Metabolism, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,Department of Laboratory Medicine, DIBIMIS, University of Palermo, Italy
| | - Luca Lovrecic
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Keli Hocevar
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tanja Papler Burnik
- Division of Obstetrics and Gynecology, Department of Human Reproduction, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Janez
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Mojca Jensterle
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Eda Vrtacnik Bokal
- Division of Obstetrics and Gynecology, Department of Human Reproduction, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Borut Peterlin
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Ales Maver
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
4
|
Obesity Animal Models for Acupuncture and Related Therapy Research Studies. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6663397. [PMID: 34630614 PMCID: PMC8497105 DOI: 10.1155/2021/6663397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 09/02/2021] [Indexed: 11/17/2022]
Abstract
Obesity and related diseases are considered as pandemic representing a worldwide threat for health. Animal models are critical to validate the effects and understand the mechanisms related to classical or innovative preventive and therapeutic strategies. It is, therefore, important to identify the best animal models for translational research, using different evaluation criteria such as the face, construct, and predictive validity. Because the pharmacological treatments and surgical interventions currently used for treating obesity often present many undesirable side effects, relatively high relapse probabilities, acupuncture, electroacupuncture (EA), and related therapies have gained more popularity and attention. Many kinds of experimental animal models have been used for obesity research studies, but in the context of acupuncture, most of the studies were performed in rodent obesity models. Though, are these obesity rodent models really the best for acupuncture or related therapies research studies? In this study, we review different obesity animal models that have been used over the past 10 years for acupuncture and EA research studies. We present their respective advantages, disadvantages, and specific constraints. With the development of research on acupuncture and EA and the increasing interest regarding these approaches, proper animal models are critical for preclinical studies aiming at developing future clinical trials in the human. The aim of the present study is to provide researchers with information and guidance related to the preclinical models that are currently available to investigate the outcomes of acupuncture and related therapies.
Collapse
|
5
|
Baranowski BJ, Allen MD, Nyarko JN, Rector RS, Padilla J, Mousseau DD, Rau CD, Wang Y, Laughlin MH, Emter CA, MacPherson RE, Olver TD. Cerebrovascular insufficiency and amyloidogenic signaling in Ossabaw swine with cardiometabolic heart failure. JCI Insight 2021; 6:143141. [PMID: 34027891 PMCID: PMC8262360 DOI: 10.1172/jci.insight.143141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 04/14/2021] [Indexed: 12/26/2022] Open
Abstract
Individuals with heart failure (HF) frequently present with comorbidities, including obesity, insulin resistance, hypertension, and dyslipidemia. Many patients with HF experience cardiogenic dementia, yet the pathophysiology of this disease remains poorly understood. Using a swine model of cardiometabolic HF (Western diet+aortic banding; WD-AB), we tested the hypothesis that WD-AB would promote a multidementia phenotype involving cerebrovascular dysfunction alongside evidence of Alzheimer’s disease (AD) pathology. The results provide evidence of cerebrovascular insufficiency coupled with neuroinflammation and amyloidosis in swine with experimental cardiometabolic HF. Although cardiac ejection fraction was normal, indices of arterial compliance and cerebral blood flow were reduced, and cerebrovascular regulation was impaired in the WD-AB group. Cerebrovascular dysfunction occurred concomitantly with increased MAPK signaling and amyloidogenic processing (i.e., increased APP, BACE1, CTF, and Aβ40 in the prefrontal cortex and hippocampus) in the WD-AB group. Transcriptomic profiles of the stellate ganglia revealed the WD-AB group displayed an enrichment of gene networks associated with MAPK/ERK signaling, AD, frontotemporal dementia, and a number of behavioral phenotypes implicated in cognitive impairment. These provide potentially novel evidence from a swine model that cerebrovascular and neuronal pathologies likely both contribute to the dementia profile in a setting of cardiometabolic HF.
Collapse
Affiliation(s)
- Bradley J Baranowski
- Department of Health Sciences and.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - Matti D Allen
- Department of Physical Medicine and Rehabilitation, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jennifer Nk Nyarko
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Darrell D Mousseau
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Christoph D Rau
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yibin Wang
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Rebecca Ek MacPherson
- Department of Health Sciences and.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
6
|
Kelty TJ, Brown JD, Kerr NR, Roberts MD, Childs TE, Cabrera OH, Manzella FM, Miller DK, Taylor GT, Booth FW. RNA-sequencing and behavioral testing reveals inherited physical inactivity co-selects for anxiogenic behavior without altering depressive-like behavior in Wistar rats. Neurosci Lett 2021; 753:135854. [PMID: 33785378 DOI: 10.1016/j.neulet.2021.135854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 11/15/2022]
Abstract
Physical inactivity is positively associated with anxiety and depression. Considering physical inactivity, anxiety, and depression each have a genetic basis for inheritance, our lab used artificial selectively bred low-voluntary running (LVR) and wild type (WT) female Wistar rats to test if physical inactivity genes selected over multiple generations would lead to an anxiety or depressive-like phenotype. We performed next generation RNA sequencing and immunoblotting on the dentate gyrus to reveal key biological functions from heritable physical inactivity. LVR rats did not display depressive-like behavior. However, LVR rats did display anxiogenic behavior with gene networks associated with reduced neuronal development, proliferation, and function compared to WT counterparts. Additionally, immunoblotting revealed LVR deficits in neuronal development and function. To our knowledge, this is the first study to show that by selectively breeding for physical inactivity genes, anxiety-like genes were co-selected. The study also reveals molecular insights to the genetic influences that physical inactivity has on anxiety-like behavior.
Collapse
Affiliation(s)
- Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA.
| | - Jacob D Brown
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Nathan R Kerr
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Michael D Roberts
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Tom E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Omar H Cabrera
- Department of Psychological Sciences, University of Missouri-St. Louis, St. Louis, MO 63110, USA
| | - Francesca M Manzella
- Department of Psychological Sciences, University of Missouri-St. Louis, St. Louis, MO 63110, USA
| | - Dennis K Miller
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - George T Taylor
- Department of Psychological Sciences, University of Missouri-St. Louis, St. Louis, MO 63110, USA
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
7
|
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 2021; 53:99-115. [PMID: 33491589 PMCID: PMC7988741 DOI: 10.1152/physiolgenomics.00093.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) patients with deteriorating right ventricular (RV) structure and function have a nearly twofold increased risk of death compared with those without. Despite the well-established clinical risk, few studies have examined the molecular signature associated with this HF condition. The purpose of this study was to integrate morphological, molecular, and functional data with the transcriptome data set in the RV of a preclinical model of cardiometabolic HF. Ossabaw swine were fed either normal diet without surgery (lean control, n = 5) or Western diet and aortic-banding (WD-AB; n = 4). Postmortem RV weight was increased and positively correlated with lung weight in the WD-AB group compared with CON. Total RNA-seq was performed and gene expression profiles were compared and analyzed using principal component analysis, weighted gene co-expression network analysis, module enrichment analysis, and ingenuity pathway analysis. Gene networks specifically associated with RV hypertrophic remodeling identified a hub gene in MAPK8 (or JNK1) that was associated with the selective induction of the extracellular matrix (ECM) component fibronectin. JNK1 and fibronectin protein were increased in the right coronary artery (RCA) of WD-AB animals and associated with a decrease in matrix metalloproteinase 14 protein, which specifically degrades fibronectin. RCA fibronectin content was correlated with increased vascular stiffness evident as a decreased elastin elastic modulus in WD-AB animals. In conclusion, this study establishes a molecular and transcriptome signature in the RV using Ossabaw swine with cardiometabolic HF. This signature was associated with altered ECM regulation and increased vascular stiffness in the RCA, with selective dysregulation of fibronectin.
Collapse
Affiliation(s)
- Shannon C Kelly
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Christoph D Rau
- Department of Computational Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - An Ouyang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pamela K Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jenna C Edwards
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Laurel A Grisanti
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Bradley S Fleenor
- Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, Indiana
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, California
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine-Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial VA Hospital, University of Missouri, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
8
|
Silva KAS, Emter CA. Large Animal Models of Heart Failure: A Translational Bridge to Clinical Success. JACC Basic Transl Sci 2020; 5:840-856. [PMID: 32875172 PMCID: PMC7452204 DOI: 10.1016/j.jacbts.2020.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
Preclinical large animal models play a critical and expanding role in translating basic science findings to the development and clinical approval of novel cardiovascular therapeutics. This state-of-the-art review outlines existing methodologies and physiological phenotypes of several HF models developed in large animals. A comprehensive list of porcine, ovine, and canine models of disease are presented, and the translational importance of these studies to clinical success is highlighted through a brief overview of recent devices approved by the FDA alongside associated clinical trials and preclinical animal reports. Increasing the use of large animal models of HF holds significant potential for identifying new mechanisms underlying this disease and providing valuable information regarding the safety and efficacy of new therapies, thus, improving physiological and economical translation of animal research to the successful treatment of human HF.
Preclinical large animal models of heart failure (HF) play a critical and expanding role in translating basic science findings to the development and clinical approval of novel therapeutics and devices. The complex combination of cardiovascular events and risk factors leading to HF has proved challenging for the development of new treatments for these patients. This state-of-the-art review presents historical and recent studies in porcine, ovine, and canine models of HF and outlines existing methodologies and physiological phenotypes. The translational importance of large animal studies to clinical success is also highlighted with an overview of recent devices approved by the Food and Drug Administration, together with preclinical HF animal studies used to aid both development and safety and/or efficacy testing. Increasing the use of large animal models of HF holds significant potential for identifying the novel mechanisms underlying the clinical condition and to improving physiological and economical translation of animal research to successfully treat human HF.
Collapse
Key Words
- AF, atrial fibrillation
- ECM, extracellular matrix
- EDP, end-diastolic pressure
- EF, ejection fraction
- FDA, Food and Drug Administration
- HF, heart failure
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- HFrEF
- HFrEF, heart failure with reduced ejection fraction
- I/R, ischemia/reperfusion
- IABP, intra-aortic balloon pump
- LAD, left anterior descending
- LCx, left circumflex
- LV, left ventricular
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- RV, right ventricular
- heart failure
- large animal model
- preclinical
Collapse
Affiliation(s)
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
9
|
Little R, Houghton MJ, Carr IM, Wabitsch M, Kerimi A, Williamson G. The Ability of Quercetin and Ferulic Acid to Lower Stored Fat is Dependent on the Metabolic Background of Human Adipocytes. Mol Nutr Food Res 2020; 64:e2000034. [PMID: 32350998 DOI: 10.1002/mnfr.202000034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/12/2020] [Indexed: 12/15/2022]
Abstract
SCOPE Dietary flavonoids and phenolic acids can modulate lipid metabolism, but effects on mature human adipocytes are not well characterized. MATERIALS AND METHODS Human adipocytes are differentiated, and contain accumulated lipids, mimicking white adipocytes. They are then cultured either under conditions of actively synthesizing and accumulating additional lipids through lipogenesis ("ongoing lipogenic state") or under conditions of maintaining but not increasing stored lipids ("lipid storage state"). Total lipid, lipidomic and transcriptomics analyses are employed to assess changes after treatment with quercetin and/or ferulic acid. RESULTS In the "lipid storage state," a longer-term treatment (3 doses over 72 h) with low concentrations of quercetin and ferulic acid together significantly lowered stored lipid content, modified lipid composition, and modulated genes related to lipid metabolism with a strong implication of peroxisome proliferator-activated receptor (PPARα)/retinoid X receptor (RXRα) involvement. In the "ongoing lipogenic state," the effect of quercetin and ferulic acid is markedly different, with fewer changes in gene expression and lipid composition, and no detectable involvement of PPARα/RXRα, with a tenfold higher concentration required to attenuate stored lipid content. CONCLUSIONS Multiple low-dose treatment of quercetin and ferulic acid modulates lipid metabolism in adipocytes, but the effect is dramatically dependent on the metabolic state of the cell.
Collapse
Affiliation(s)
- Robert Little
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Michael J Houghton
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.,Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | - Ian M Carr
- Saint James' University Hospital, Granville Road, Leeds, LS9 7TF, UK
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine University Medical Centre, University of Ulm, Ulm, 89075, Germany
| | - Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.,Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.,Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| |
Collapse
|
10
|
Ballester M, Quintanilla R, Ortega FJ, Serrano JCE, Cassanyé A, Rodríguez-Palmero M, Moreno-Muñoz JA, Portero-Otin M, Tibau J. Dietary intake of bioactive ingredients impacts liver and adipose tissue transcriptomes in a porcine model of prepubertal early obesity. Sci Rep 2020; 10:5375. [PMID: 32214182 PMCID: PMC7096439 DOI: 10.1038/s41598-020-62320-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
Global prevalence of obesity has increased to epidemic proportions over the past 40 years, with childhood obesity reaching alarming rates. In this study, we determined changes in liver and adipose tissue transcriptomes of a porcine model for prepubertal early obesity induced by a high-calorie diet and supplemented with bioactive ingredients. A total of 43 nine-weeks-old animals distributed in four pens were fed with four different dietary treatments for 10 weeks: a conventional diet; a western-type diet; and a western-type diet with Bifidobacterium breve and rice hydrolysate, either adding or not omega-3 fatty acids. Animals fed a western-type diet increased body weight and total fat content and exhibited elevated serum concentrations of cholesterol, whereas animals supplemented with bioactive ingredients showed lower body weight gain and tended to accumulate less fat. An RNA-seq experiment was performed with a total of 20 animals (five per group). Differential expression analyses revealed an increase in lipogenesis, cholesterogenesis and inflammatory processes in animals on the western-type diet while the supplementation with bioactive ingredients induced fatty acid oxidation and cholesterol catabolism, and decreased adipogenesis and inflammation. These results reveal molecular mechanisms underlying the beneficial effects of bioactive ingredient supplementation in an obese pig model.
Collapse
Affiliation(s)
- Maria Ballester
- Animal Breeding and Genetics Programme, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140, Caldes de Montbui, Spain.
| | - Raquel Quintanilla
- Animal Breeding and Genetics Programme, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140, Caldes de Montbui, Spain
| | - Francisco J Ortega
- Department of Diabetes, Endocrinology, and Nutrition (UDEN), Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José C E Serrano
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25196, Lleida, Spain
| | - Anna Cassanyé
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25196, Lleida, Spain
| | | | | | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25196, Lleida, Spain
| | - Joan Tibau
- Animal Breeding and Genetics Programme, Institute for Research and Technology in Food and Agriculture (IRTA), Finca Camps i Armet, 17121, Monells, Spain
| |
Collapse
|
11
|
Swanson K, Kutzler M, Bionaz M. Cow milk does not affect adiposity in growing piglets as a model for children. J Dairy Sci 2019; 102:4798-4807. [PMID: 30904312 DOI: 10.3168/jds.2018-15201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
Abstract
The effect of milk consumption on childhood obesity is unclear and a direct demonstration of an association is needed. In the present study, we used piglets as a model for prepubertal children to determine the effect of milk on adipose tissue. Two studies were conducted: study 1 with 5-wk-old male piglets (n = 8) and study 2 with 8- to 9-wk-old male piglets (n = 12). The piglets were fed a normal growing diet and randomly assigned to receive daily either 750 mL of whole cow milk or an isocaloric maltodextrin solution (control). For approximately 12 wk, body weight, feed intake, and subcutaneous back fat thickness were determined ultrasonographically and recorded. At euthanasia, back and neck fat thicknesses were measured and samples of back fat were collected for adipose histology. In study 1, but not study 2, piglets receiving milk grew more and ate more compared with control. In study 1, both back fat and neck fat thickness were greater in the milk-fed piglets and they had a higher frequency of small adipocytes and a lower frequency of intermediate and large adipocytes compared with controls. In study 2, control pigs had a significantly greater frequency of intermediate adipocytes but the milk-fed piglets tended to have a higher frequency of the largest adipocytes. In conclusion, milk has no apparent causal or consistent effect on adipose tissue in growing piglets.
Collapse
Affiliation(s)
- Katherine Swanson
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331
| | - Michelle Kutzler
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331.
| |
Collapse
|
12
|
Liu X, Gong J, Wang L, Hou X, Gao H, Yan H, Zhao F, Zhang L, Wang L. Genome-Wide Profiling of the Microrna Transcriptome Regulatory Network to Identify Putative Candidate Genes Associated with Backfat Deposition in Pigs. Animals (Basel) 2019; 9:ani9060313. [PMID: 31159441 PMCID: PMC6617047 DOI: 10.3390/ani9060313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/27/2019] [Accepted: 05/30/2019] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Backfat thickness is an important characteristic in pig breeding. In this study, the key microRNAs (miRNAs) and genes associated with pig backfat deposition were detected and characterized using RNA sequencing between adipose tissues of high-backfat and low-backfat pigs. Strong candidate mRNA‒miRNA interaction pairs were identified to affect backfat deposition through the regulation of target genes by miRNAs. These results provide novel insights into the backfat deposition mechanism in pigs. Abstract Backfat deposition is strongly related to carcass traits, growth rate, feed conversion rate, and reproductive performance in pig production. To understand the molecular mechanisms underlying porcine backfat thickness phenotypes, transcriptome and miRNA profiling of backfat from high-backfat thickness and low-backfat thickness pigs were performed by RNA sequencing. Twenty genes encoding for miRNAs and 126 genes encoding for protein-coding genes were found to be differentially expressed between the two libraries. After integrative analysis of DEMs targets and DEGs, a total of 33 mRNA‒miRNA interaction pairs were identified, and the regulatory networks of these pairs were determined. Among these genes, five (AQP9, DKK3, GLYCTK, GLIPR1, and DUSP2) related to fat deposition were found to be strong candidate genes, and mir-31-5p/AQP9 and mir-31-5p/GLIPR1 may play important roles in fat deposition. Additionally, potential adipogenesis-related genes and miRNAs were identified. These findings improve the current understanding of the molecular genetic mechanisms of subcutaneous fat deposition in pigs and provide a foundation for further studies.
Collapse
Affiliation(s)
- Xin Liu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Jianfei Gong
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China.
| | - Ligang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xinhua Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Hongmei Gao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Hua Yan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Fuping Zhao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Longchao Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
13
|
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 2019; 4:404-421. [PMID: 31312763 PMCID: PMC6610000 DOI: 10.1016/j.jacbts.2019.02.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
Collapse
Key Words
- AB, aortic-banded
- CON, control
- EDPVR, end-diastolic pressure−volume relationship
- EF, ejection fraction
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL1RL1, interleukin 1 receptor-like 1
- LV, left ventricle
- NF, nuclear factor
- PTX3, pentraxin-3
- WD, Western Diet
- cardio-metabolic disease
- heart failure
- integrative pathophysiology
- preclinical model of cardiovascular disease
Collapse
Affiliation(s)
- T. Dylan Olver
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Jenna C. Edwards
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Thomas J. Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - John L. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Chen Gao
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christoph Rau
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chad M. Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Paula J. Klutho
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Linda Alex
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | | | - Jan R. Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Pamela K. Thorne
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Christopher P. Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A. Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University- School of Medicine, St. Louis, Missouri
| | - Timothy L. Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
- Department of Child Health, University of Missouri-Columbia, Columbia, Missouri
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Department of Medicine – University of Missouri-Columbia, Columbia, Missouri
- Research Service, Harry S Truman Memorial VA Hospital, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A. Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
14
|
Walker ME, Matthan NR, Lamon-Fava S, Solano-Aguilar G, Jang S, Lakshman S, Molokin A, Urban JF, Faits T, Johnson WE, Lichtenstein AH. A Western-Type Dietary Pattern Induces an Atherogenic Gene Expression Profile in the Coronary Arteries of the Ossabaw Pig. Curr Dev Nutr 2019; 3:nzz023. [PMID: 31049488 PMCID: PMC6488722 DOI: 10.1093/cdn/nzz023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/27/2019] [Accepted: 03/28/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Current cardiovascular risk reduction guidance focuses on shifts in dietary patterns, rather than single foods or nutrients. Experimental studies are needed to identify the mechanisms by which food-based diets affect the development and progression of atherosclerosis. OBJECTIVES The aim of this study was to investigate the effect of 2 food-based dietary patterns and statin therapy on the transcriptome of the left anterior descending coronary artery of the Ossabaw pig. METHODS Pigs were randomly assigned to 1 of 4 groups and fed isocaloric diets for 6 mo; Heart Healthy-style diet (HHD) (high in unsaturated fat, unrefined grain, fruits/vegetables) or Western-style diet (WD) (high in saturated fat, cholesterol, refined grain), with or without atorvastatin. A 2-factor edge R analysis was used to determine differential gene expression in the left anterior descending coronary artery. RESULTS Relative to the HHD, the WD resulted in the differential expression of 143 genes, of which 139 genes were upregulated and 4 genes were downregulated (all log fold change ≥0.6, false discovery rate <0.10). The WD, compared with the HHD, resulted in the statistically significant upregulation of 8 atherosclerosis-associated pathways implicated in immune and inflammatory processes. There were no genes with significant differential expression attributable to statin therapy. CONCLUSIONS These data suggest that a WD induces alterations in the transcriptome of the coronary artery consistent with an inflammatory atherogenic phenotype in the Ossabaw pig with no significant modification by concurrent statin therapy.
Collapse
Affiliation(s)
- Maura E Walker
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Gloria Solano-Aguilar
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Saebyeol Jang
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Sukla Lakshman
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Aleksey Molokin
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Joseph F Urban
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Tyler Faits
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - W Evan Johnson
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Alice H Lichtenstein
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| |
Collapse
|
15
|
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 2019; 19:884-894. [PMID: 30387338 DOI: 10.1631/jzus.b1700507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Because of their physiological similarity to humans, pigs provide an excellent model for the study of obesity. This study evaluated diet-induced adiposity in genetically lean pigs and found that body weight and energy intake did not differ between controls and pigs fed the high-fat (HF) diet for three months. However, fat mass percentage, adipocyte size, concentrations of total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C), insulin, and leptin in plasma were significantly higher in HF pigs than in controls. The HF diet increased the expression in backfat tissue of genes responsible for cholesterol synthesis such as Insig-1 and Insig-2. Lipid metabolism-related genes including sterol regulatory element binding protein 1c (SREBP-1c), fatty acid synthase 1 (FASN1), diacylglycerol O-acyltransferase 2 (DGAT2), and fatty acid binding protein 4 (FABP4) were significantly up-regulated in backfat tissue, while the expression of proliferator-activated receptor-α (PPAR-α) and carnitine palmitoyl transferase 2 (CPT2), both involved in fatty acid oxidation, was reduced. In liver tissue, HF feeding significantly elevated the expression of SREBP-1c, FASN1, DGAT2, and hepatocyte nuclear factor-4α (HNF-4α) mRNAs. Microarray analysis further showed that the HF diet had a significant effect on the expression of 576 genes. Among these, 108 genes were related to 21 pathways, with 20 genes involved in adiposity deposition and 26 related to immune response. Our results suggest that an HF diet can induce genetically lean pigs into obesity with body fat mass expansion and adipose-related inflammation.
Collapse
Affiliation(s)
- Xue-Fen Yang
- Laboratory of Animal Nutrition and Feed (South China), Ministry of Agriculture / State Key Laboratory of Livestock and Poultry Breeding / Guangdong Key Laboratory of Animal Breeding and Nutrition / Guangdong Public Laboratory of Animal Breeding and Nutrition / Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Yue-Qin Qiu
- Laboratory of Animal Nutrition and Feed (South China), Ministry of Agriculture / State Key Laboratory of Livestock and Poultry Breeding / Guangdong Key Laboratory of Animal Breeding and Nutrition / Guangdong Public Laboratory of Animal Breeding and Nutrition / Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Li Wang
- Laboratory of Animal Nutrition and Feed (South China), Ministry of Agriculture / State Key Laboratory of Livestock and Poultry Breeding / Guangdong Key Laboratory of Animal Breeding and Nutrition / Guangdong Public Laboratory of Animal Breeding and Nutrition / Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Kai-Guo Gao
- Laboratory of Animal Nutrition and Feed (South China), Ministry of Agriculture / State Key Laboratory of Livestock and Poultry Breeding / Guangdong Key Laboratory of Animal Breeding and Nutrition / Guangdong Public Laboratory of Animal Breeding and Nutrition / Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Zong-Yong Jiang
- Laboratory of Animal Nutrition and Feed (South China), Ministry of Agriculture / State Key Laboratory of Livestock and Poultry Breeding / Guangdong Key Laboratory of Animal Breeding and Nutrition / Guangdong Public Laboratory of Animal Breeding and Nutrition / Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| |
Collapse
|
16
|
Starbæk SMR, Brogaard L, Dawson HD, Smith AD, Heegaard PMH, Larsen LE, Jungersen G, Skovgaard K. Animal Models for Influenza A Virus Infection Incorporating the Involvement of Innate Host Defenses: Enhanced Translational Value of the Porcine Model. ILAR J 2018; 59:323-337. [PMID: 30476076 DOI: 10.1093/ilar/ily009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 06/19/2018] [Indexed: 01/05/2025] Open
Abstract
Influenza is a viral respiratory disease having a major impact on public health. Influenza A virus (IAV) usually causes mild transitory disease in humans. However, in specific groups of individuals such as severely obese, the elderly, and individuals with underlying inflammatory conditions, IAV can cause severe illness or death. In this review, relevant small and large animal models for human IAV infection, including the pig, ferret, and mouse, are discussed. The focus is on the pig as a large animal model for human IAV infection as well as on the associated innate immune response. Pigs are natural hosts for the same IAV subtypes as humans, they develop clinical disease mirroring human symptoms, they have similar lung anatomy, and their respiratory physiology and immune responses to IAV infection are remarkably similar to what is observed in humans. The pig model shows high face and target validity for human IAV infection, making it suitable for modeling many aspects of influenza, including increased risk of severe disease and impaired vaccine response due to underlying pathologies such as low-grade inflammation. Comparative analysis of proteins involved in viral pattern recognition, interferon responses, and regulation of interferon-stimulated genes reveals a significantly higher degree of similarity between pig, ferret, and human compared with mice. It is concluded that the pig is a promising animal model displaying substantial human translational value with the ability to provide essential insights into IAV infection, pathogenesis, and immunity.
Collapse
Affiliation(s)
- Sofie M R Starbæk
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Louise Brogaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Harry D Dawson
- Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland
| | - Allen D Smith
- Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland
| | - Peter M H Heegaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars E Larsen
- National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
17
|
Jurrissen TJ, Olver TD, Winn NC, Grunewald ZI, Lin GS, Hiemstra JA, Edwards JC, Gastecki ML, Welly RJ, Emter CA, Vieira-Potter, VJ, Padilla J. Endothelial dysfunction occurs independently of adipose tissue inflammation and insulin resistance in ovariectomized Yucatan miniature-swine. Adipocyte 2018; 7:35-44. [PMID: 29283284 DOI: 10.1080/21623945.2017.1405191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
In rodents, experimentally-induced ovarian hormone deficiency increases adiposity and adipose tissue (AT) inflammation, which is thought to contribute to insulin resistance and increased cardiovascular disease risk. However, whether this occurs in a translationally-relevant large animal model remains unknown. Herein, we tested the hypothesis that ovariectomy would promote visceral and perivascular AT (PVAT) inflammation, as well as subsequent insulin resistance and peripheral vascular dysfunction in female swine. At sexual maturity (7 months of age), female Yucatan mini-swine either remained intact (control, n = 9) or were ovariectomized (OVX, n = 7). All pigs were fed standard chow (15-20 g/kg), and were euthanized 6 months post-surgery. Uterine mass and plasma estradiol levels were decreased by ∼10-fold and 2-fold, respectively, in OVX compared to control pigs. Body mass, glucose homeostasis, and markers of insulin resistance were not different between control and OVX pigs; however, OVX animals exhibited greater plasma triglycerides and triglyceride:HDL ratio. Ovariectomy enhanced visceral adipocyte expansion, although this was not accompanied by brachial artery PVAT adipocyte expansion, AT inflammation in either depot, or increased systemic inflammation assessed by plasma C-reactive protein concentrations. Despite the lack of AT inflammation and insulin resistance, OVX pigs exhibited depressed brachial artery endothelial-dependent vasorelaxation, which was rescued with blockade of endothelin receptor A. Together, these findings indicate that in female Yucatan mini-swine, increased AT inflammation and insulin resistance are not required for loss of ovarian hormones to induce endothelial dysfunction.
Collapse
Affiliation(s)
- Thomas J. Jurrissen
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - T. Dylan Olver
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Nathan C. Winn
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Zachary I. Grunewald
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Gabriela S. Lin
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Biology, Barry University, Miami, FL, United States
| | | | - Jenna C. Edwards
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Michelle L. Gastecki
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Rebecca J. Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | | | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Child Health, University of Missouri, Columbia, MO, United States
| |
Collapse
|
18
|
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 Metab 2018; 314:E78-E92. [PMID: 28899857 PMCID: PMC5866386 DOI: 10.1152/ajpendo.00015.2017] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Pediatric obesity and nonalcoholic steatohepatitis (NASH) are on the rise in industrialized countries, yet our ability to mechanistically examine this relationship is limited by the lack of a suitable higher animal models. Here, we examined the effects of high-fat, high-fructose corn syrup, high-cholesterol Western-style diet (WD)-induced obesity on NASH and cecal microbiota dysbiosis in juvenile Ossabaw swine. Juvenile female Ossabaw swine (5 wk old) were fed WD (43.0% fat; 17.8% high-fructose corn syrup; 2% cholesterol) or low-fat diet (CON/lean; 10.5% fat) for 16 wk ( n = 6 each) or 36 wk ( n = 4 each). WD-fed pigs developed obesity, dyslipidemia, and systemic insulin resistance compared with CON pigs. In addition, obese WD-fed pigs developed severe NASH, with hepatic steatosis, hepatocyte ballooning, inflammatory cell infiltration, and fibrosis after 16 wk, with further exacerbation of histological inflammation and fibrosis after 36 wk of WD feeding. WD feeding also resulted in robust cecal microbiota changes including increased relative abundances of families and genera in Proteobacteria ( P < 0.05) (i.e., Enterobacteriaceae, Succinivibrionaceae, and Succinivibrio) and LPS-containing Desulfovibrionaceae and Desulfovibrio and a greater ( P < 0.05) predicted microbial metabolic function for LPS biosynthesis, LPS biosynthesis proteins, and peptidoglycan synthesis compared with CON-fed pigs. Overall, juvenile Ossabaw swine fed a high-fat, high-fructose, high-cholesterol diet develop obesity and severe microbiota dysbiosis with a proinflammatory signature and a NASH phenotype directly relevant to the pediatric/adolescent and young adult population.
Collapse
Affiliation(s)
- M. R. Panasevich
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - G. M. Meers
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - M. A. Linden
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - F. W. Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - J. W. Perfield
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Food Science, University of Missouri, Columbia, Missouri
| | - K. L. Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Umesh D. Wankhade
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sree V. Chintapalli
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - K. Shankar
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - J. A. Ibdah
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - R. S. Rector
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
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 2017; 314:R252-R264. [PMID: 29141949 DOI: 10.1152/ajpregu.00213.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Impaired microvascular insulin signaling may develop before overt indices of microvascular endothelial dysfunction and represent an early pathological feature of adolescent obesity. Using a translational porcine model of juvenile obesity, we tested the hypotheses that in the early stages of obesity development, impaired insulin signaling manifests in skeletal muscle (triceps), brain (prefrontal cortex), and corresponding vasculatures, and that depressed insulin-induced vasodilation is reversible with acute inhibition of protein kinase Cβ (PKCβ). Juvenile Ossabaw miniature swine (3.5 mo of age) were divided into two groups: lean control ( n = 6) and obese ( n = 6). Obesity was induced by feeding the animals a high-fat/high-fructose corn syrup/high-cholesterol diet for 10 wk. Juvenile obesity was characterized by excess body mass, hyperglycemia, physical inactivity (accelerometer), and marked lipid accumulation in the skeletal muscle, with no evidence of overt atherosclerotic lesions in athero-prone regions, such as the abdominal aorta. Endothelium-dependent (bradykinin) and -independent (sodium nitroprusside) vasomotor responses in the brachial and carotid arteries (wire myography), as well as in the skeletal muscle resistance and 2A pial arterioles (pressure myography) were unaltered, but insulin-induced microvascular vasodilation was impaired in the obese group. Blunted insulin-stimulated vasodilation, which was reversed with acute PKCβ inhibition (LY333-531), occurred alongside decreased tissue perfusion, as well as reduced insulin-stimulated Akt signaling in the prefrontal cortex, but not the triceps. In the early stages of juvenile obesity development, the microvasculature and prefrontal cortex exhibit impaired insulin signaling. Such adaptations may underscore vascular and neurological derangements associated with juvenile obesity.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri , Columbia, Missouri
| | - Zachary I Grunewald
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Thomas J Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | | | - Paul J LeBlanc
- Department of Health Sciences, Brock University , St. Catharines, Ontario , Canada
| | - Teagan R Schnurbusch
- National Swine Resource and Research Center University of Missouri , Columbia, Missouri
| | - Alana M Czajkowski
- National Swine Resource and Research Center University of Missouri , Columbia, Missouri
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri , Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Affairs Hospital , Columbia, Missouri.,Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri , Columbia, Missouri
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri , Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Affairs Hospital , Columbia, Missouri
| | - Eric M Walters
- National Swine Resource and Research Center University of Missouri , Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri , Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Department of Child Health, University of Missouri , Columbia, Missouri
| |
Collapse
|
20
|
Martin JS, Kephart WC, Haun CT, McCloskey AE, Shake JJ, Mobley CB, Goodlett MD, Kavazis A, Pascoe DD, Zhang L, Roberts MD. Impact of external pneumatic compression target inflation pressure on transcriptome-wide RNA expression in skeletal muscle. Physiol Rep 2017; 4:4/22/e13029. [PMID: 27884954 PMCID: PMC5357997 DOI: 10.14814/phy2.13029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/03/2016] [Accepted: 10/12/2016] [Indexed: 11/24/2022] Open
Abstract
Next‐generation RNA sequencing was employed to determine the acute and subchronic impact of peristaltic pulse external pneumatic compression (PEPC) of different target inflation pressures on global gene expression in human vastus lateralis skeletal muscle biopsy samples. Eighteen (N = 18) male participants were randomly assigned to one of the three groups: (1) sham (n = 6), 2) EPC at 30–40 mmHg (LP‐EPC; n = 6), and 3) EPC at 70–80 mmHg (MP‐EPC; n = 6). One hour treatment with sham/EPC occurred for seven consecutive days. Vastus lateralis skeletal muscle biopsies were performed at baseline (before first treatment; PRE), 1 h following the first treatment (POST1), and 24 h following the last (7th) treatment (POST2). Changes from PRE in gene expression were analyzed via paired comparisons within each group. Genes were filtered to include only those that had an RPKM ≥ 1.0, a fold‐change of ≥1.5 and a paired t‐test value of <0.01. For the sham condition, two genes at POST1 and one gene at POST2 were significantly altered. For the LP‐EPC condition, nine genes were up‐regulated and 0 genes were down‐regulated at POST1 while 39 genes were up‐regulated and one gene down‐regulated at POST2. For the MP‐EPC condition, two genes were significantly up‐regulated and 21 genes were down‐regulated at POST1 and 0 genes were altered at POST2. Both LP‐EPC and MP‐EPC acutely alter skeletal muscle gene expression, though only LP‐EPC appeared to affect gene expression with subchronic application. Moreover, the transcriptome response to EPC demonstrated marked heterogeneity (i.e., genes and directionality) with different target inflation pressures.
Collapse
Affiliation(s)
- Jeffrey S Martin
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama .,School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Cody T Haun
- School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Joshua J Shake
- School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Michael D Goodlett
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,Athletics Department, Auburn University, Auburn, Alabama
| | - Andreas Kavazis
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,School of Kinesiology, Auburn University, Auburn, Alabama
| | - David D Pascoe
- School of Kinesiology, Auburn University, Auburn, Alabama
| | - Lee Zhang
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama
| | - Michael D Roberts
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,School of Kinesiology, Auburn University, Auburn, Alabama
| |
Collapse
|
21
|
Panasevich MR, Peppler WT, Oerther DB, Wright DC, Rector RS. Microbiome and NAFLD: potential influence of aerobic fitness and lifestyle modification. Physiol Genomics 2017; 49:385-399. [PMID: 28600319 DOI: 10.1152/physiolgenomics.00012.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease with prevalence rates that are on the rise in the US and worldwide. NAFLD encompasses a spectrum of liver pathologies including simple steatosis to nonalcoholic steatohepatitis (NASH) with inflammation and fibrosis. The gut microbiome has emerged as a potential therapeutic target in combating metabolic diseases including obesity, Type 2 diabetes, and NAFLD/NASH. Diet-induced obesity/Western style diet feeding causes severe microbial dysbiosis initiating a microbiome signature that promotes metabolite production that directly impacts hepatic metabolism. Changes in lifestyle (i.e., diet, exercise, and aerobic fitness) improve NAFLD outcomes and can significantly influence the microbiome. However, directly linking lifestyle-induced remodeling of the microbiome to NAFLD pathogenesis is not well understood. Understanding the reshaping of the microbiome and the metabolites produced and their subsequent actions on hepatic metabolism are vital in understanding the gut-liver axis. In this review, we 1) discuss microbiome-derived metabolites that significantly contribute to the gut-liver axis and are directly linked to NAFLD/NASH and 2) present evidence on lifestyle modifications reshaping the microbiome and the potential therapeutic aspects in combating the disease.
Collapse
Affiliation(s)
- Matthew R Panasevich
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri.,Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology; University of Missouri, Columbia, Missouri
| | - Willem T Peppler
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Daniel B Oerther
- Department of Civil, Architectural, and Environmental Engineering, Missouri University of Science and Technology, Rolla, Missouri; and
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - R Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri; .,Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology; University of Missouri, Columbia, Missouri
| |
Collapse
|
22
|
Mentzel CMJ, Cardoso TF, Lex AMJ, Sørensen DB, Fredholm M, Cirera S. Fat and carbohydrate content in the diet induces drastic changes in gene expression in young Göttingen minipigs. Mamm Genome 2017; 28:166-175. [PMID: 28396939 DOI: 10.1007/s00335-017-9690-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/04/2017] [Indexed: 12/20/2022]
Abstract
In human health, there is interest in developing specific diets to reduce body weight. These studies are mainly focused on phenotypic changes induced in blood measurements, i.e., triglycerides, HDL, LDL, and insulin, and on physical changes, i.e., body weight and BMI. To evaluate the biological impact of diet interventions, it is very important to investigate the molecular mechanisms driving the diet-induced phenotypic changes in relevant tissues. However, studying these effects in humans is difficult due to ethical concerns in doing interventions and obtaining tissue samples and good animal models are therefore needed. Göttingen minipigs, a small size obesity prone pig breed, have previously been shown to be a useful translational animal model for metabolic studies. In this study, 16 Göttingen minipig males (2-month old) were submitted to 13 weeks of differential diets to investigate the initial stages of diet-induced metabolic changes. Half of them were fed a high-fat/cholesterol, low-carbohydrate (HFLC) diet, and the other half were fed a low- fat/cholesterol, high-carbohydrate (LFHC) diet. After 13 weeks, the HFLC group weighted less and had dyslipidemia compared to the LFHC group. Liver, pancreas, and adipose tissues were collected at slaughter. Gene expression profiling of 83 metabolism-relevant genes was performed using high-throughput qPCR. In total, 41 genes were deregulated in at least one of the five tissues analyzed, with liver being the most drastically affected tissue. The new knowledge gained in this study could potentially be of value for considering direct modulation of gene expression by nutrient content in the diet.
Collapse
Affiliation(s)
- Caroline M Junker Mentzel
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Tainã Figueiredo Cardoso
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Department of Animal Genetics, Center for Research in Agricultural Genomics (CSIC-IRTA-UAB-UB), Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Annika M J Lex
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Dorte Bratbo Sørensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
23
|
Plassais J, Rimbault M, Williams FJ, Davis BW, Schoenebeck JJ, Ostrander EA. Analysis of large versus small dogs reveals three genes on the canine X chromosome associated with body weight, muscling and back fat thickness. PLoS Genet 2017; 13:e1006661. [PMID: 28257443 PMCID: PMC5357063 DOI: 10.1371/journal.pgen.1006661] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/17/2017] [Accepted: 02/26/2017] [Indexed: 12/21/2022] Open
Abstract
Domestic dog breeds display significant diversity in both body mass and skeletal size, resulting from intensive selective pressure during the formation and maintenance of modern breeds. While previous studies focused on the identification of alleles that contribute to small skeletal size, little is known about the underlying genetics controlling large size. We first performed a genome-wide association study (GWAS) using the Illumina Canine HD 170,000 single nucleotide polymorphism (SNP) array which compared 165 large-breed dogs from 19 breeds (defined as having a Standard Breed Weight (SBW) >41 kg [90 lb]) to 690 dogs from 69 small breeds (SBW ≤41 kg). We identified two loci on the canine X chromosome that were strongly associated with large body size at 82-84 megabases (Mb) and 101-104 Mb. Analyses of whole genome sequencing (WGS) data from 163 dogs revealed two indels in the Insulin Receptor Substrate 4 (IRS4) gene at 82.2 Mb and two additional mutations, one SNP and one deletion of a single codon, in Immunoglobulin Superfamily member 1 gene (IGSF1) at 102.3 Mb. IRS4 and IGSF1 are members of the GH/IGF1 and thyroid pathways whose roles include determination of body size. We also found one highly associated SNP in the 5'UTR of Acyl-CoA Synthetase Long-chain family member 4 (ACSL4) at 82.9 Mb, a gene which controls the traits of muscling and back fat thickness. We show by analysis of sequencing data from 26 wolves and 959 dogs representing 102 domestic dog breeds that skeletal size and body mass in large dog breeds are strongly associated with variants within IRS4, ACSL4 and IGSF1.
Collapse
Affiliation(s)
- Jocelyn Plassais
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maud Rimbault
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Falina J. Williams
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brian W. Davis
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey J. Schoenebeck
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elaine A. Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
24
|
Ruegsegger GN, Toedebusch RG, Braselton JF, Childs TE, Booth FW. Left ventricle transcriptomic analysis reveals connective tissue accumulation associates with initial age-dependent decline in V̇o2peak from its lifetime apex. Physiol Genomics 2016; 49:53-66. [PMID: 27913688 DOI: 10.1152/physiolgenomics.00083.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/08/2016] [Accepted: 11/28/2016] [Indexed: 12/17/2022] Open
Abstract
Peak oxygen consumption (V̇o2peak) strongly predicts morbidity and mortality better than other established risk factors, yet mechanisms associated with its age-associated decline are unknown. Our laboratory has shown that V̇o2peak first begins to decrease at the same age of 19-20 wk in both sedentary and wheel-running, female Wistar rats (Toedebusch et al., Physiol Genomics 48: 101-115, 2016). Here, we employed a total systemic approach using unsupervised interrogation of mRNA with RNA sequencing. The purpose of our study was to analyze transcriptomic profiles from both sedentary (SED) and wheel-running (RUN) conditions as a strategy to identify pathways in the left ventricle that may contribute to the initial reductions in V̇o2peak occurring between 19 and 27 wk of age. Transcriptomic comparisons were made within both SED and RUN rats between 19 and 27 wk (n = 5-8). Analysis of mRNAs shared in SED and RUN between 19 and 27 wk found 17 upregulated (e.g., Adra1d, Rpl17, Xpo7) and 8 downregulated (e.g., Cdo1, Ctfg, Sfrp1) mRNAs, at 19 wk, respectively. Furthermore, bioinformatics analysis of mRNAs common to SED and RUN produced networks suggestive of increased connective tissue development at 27 vs. 19 wk. Additionally, Ctfg mRNA was negatively associated with V̇o2peak in both SED and RUN (P < 0.05). In summary, transcriptomic analysis revealed mRNAs and networks associated with increased connective tissue development, decreased α-adrenergic activity, and decreased protein translation in the left ventricle that could, in part, potentially influence the initiation of the lifelong reduction in V̇o2peak, independent of physical activity levels.
Collapse
Affiliation(s)
| | - Ryan G Toedebusch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Joshua F Braselton
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Thomas E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri; .,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; and.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
25
|
Sheldon RD, Kanosky KM, Wells KD, Miles L, Perfield JW, Xanthakos S, Inge TH, Rector RS. Transcriptomic differences in intra-abdominal adipose tissue in extremely obese adolescents with different stages of NAFLD. Physiol Genomics 2016; 48:897-911. [PMID: 27764764 DOI: 10.1152/physiolgenomics.00020.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 10/04/2016] [Indexed: 02/07/2023] Open
Abstract
Mechanisms responsible for progression of nonalcoholic fatty liver disease (NAFLD) to steatohepatitis (NASH) remain poorly defined. To examine the potential contribution of adipose tissue to NAFLD progression, we performed a complete transcriptomic analysis using RNA sequencing (RNA-Seq) on intra-abdominal adipose tissue (IAT) from severely obese adolescents [Mage 16.9 ± 0.4 yr, body mass index (BMI) z-score 2.7 ± 0.1] undergoing bariatric surgery and liver biopsy categorized into three groups: no steatosis (normal, n = 8), steatosis only (n = 13), or NASH (n = 10) by liver histology. Age, body weight, and BMI did not differ among groups, but subjects with NASH were more insulin resistant (increased homeostatic model assessment/insulin resistance, P < 0.05 vs. other groups). RNA-Seq revealed 175 up- and 492 downregulated mRNA transcripts (≥±1.5-fold, false discovery rate <0.10) in IAT between NASH vs. Normal, with "mitochondrial dysfunction, P = 4.19E-7" being the top regulated canonical pathway identified by Ingenuity Pathway Analysis; only 19 mRNA transcripts were up- and 148 downregulated when comparing Steatosis vs. Normal, with suppression of "EIF2 signaling, P = 1.79E-27" being the top regulated pathway indicating increased cellular stress. A comparison of IAT between NASH vs. Steatosis found 515 up- and 175 downregulated genes, with "antigen presentation, P = 6.03E-18" being the top regulated canonical pathway and "inflammatory response" the top diseases and disorders function. Unique transcriptomic differences exist in IAT from severely obese adolescents with distinct stages of NAFLD, providing an important resource for identifying potential novel therapeutic targets for childhood NASH.
Collapse
Affiliation(s)
- Ryan D Sheldon
- Research Service, Harry S. Truman Memorial Veterans Medical Center, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Kayla M Kanosky
- Research Service, Harry S. Truman Memorial Veterans Medical Center, Columbia, Missouri.,Department of Medicine-Division of Gastroenterology and Hepatology University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Kevin D Wells
- Department of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Lili Miles
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - James W Perfield
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri.,Department of Food Sciences, University of Missouri, Columbia, Missouri; and
| | | | - Thomas H Inge
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - R Scott Rector
- Research Service, Harry S. Truman Memorial Veterans Medical Center, Columbia, Missouri; .,Department of Medicine-Division of Gastroenterology and Hepatology University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
26
|
Stachowiak M, Szczerbal I, Switonski M. Genetics of Adiposity in Large Animal Models for Human Obesity-Studies on Pigs and Dogs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:233-70. [PMID: 27288831 DOI: 10.1016/bs.pmbts.2016.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of domestic mammals in the development of human biomedical sciences has been widely documented. Among these model species the pig and dog are of special importance. Both are useful for studies on the etiology of human obesity. Genome sequences of both species are known and advanced genetic tools [eg, microarray SNP for genome wide association studies (GWAS), next generation sequencing (NGS), etc.] are commonly used in such studies. In the domestic pig the accumulation of adipose tissue is an important trait, which influences meat quality and fattening efficiency. Numerous quantitative trait loci (QTLs) for pig fatness traits were identified, while gene polymorphisms associated with these traits were also described. The situation is different in dog population. Generally, excessive accumulation of adipose tissue is considered, similar to humans, as a complex disease. However, research on the genetic background of canine obesity is still in its infancy. Between-breed differences in terms of adipose tissue accumulation are well known in both animal species. In this review we show recent advances of studies on adipose tissue accumulation in pigs and dogs, and their potential importance for studies on human obesity.
Collapse
Affiliation(s)
- M Stachowiak
- Department of Genetics, Animal Breeding, Poznań University of Life Sciences, Poznań, Poland
| | - I Szczerbal
- Department of Genetics, Animal Breeding, Poznań University of Life Sciences, Poznań, Poland
| | - M Switonski
- Department of Genetics, Animal Breeding, Poznań University of Life Sciences, Poznań, Poland.
| |
Collapse
|
27
|
Zambonelli P, Gaffo E, Zappaterra M, Bortoluzzi S, Davoli R. Transcriptional profiling of subcutaneous adipose tissue in Italian Large White pigs divergent for backfat thickness. Anim Genet 2016; 47:306-23. [PMID: 26931818 DOI: 10.1111/age.12413] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2015] [Indexed: 12/30/2022]
Abstract
Fat deposition is a widely studied trait in pigs because of its implications with animal growth efficiency, technological and nutritional characteristics of meat products, but the global framework of the biological and molecular processes regulating fat deposition in pigs is still incomplete. This study describes the backfat tissue transcription profile in Italian Large White pigs and reports genes differentially expressed between fat and lean animals according to RNA-seq data. The backfat transcription profile was characterised by the expression of 23 483 genes, of which 54.1% were represented by known genes. Of 63 418 expressed transcripts, about 80% were non-previously annotated isoforms. By comparing the expression level of fat vs. lean pigs, we detected 86 robust differentially expressed transcripts, 72 more highly expressed (e.g. ACP5, BCL2A1, CCR1, CD163, CD1A, EGR2, ENPP1, GPNMB, INHBB, LYZ, MSR1, OLR1, PIK3AP1, PLIN2, SPP1, SLC11A1, STC1) and 14 lower expressed (e.g. ADSSL1, CDO1, DNAJB1, HSPA1A, HSPA1B, HSPA2, HSPB8, IGFBP5, OLFML3) in fat pigs. The main functional categories enriched in differentially expressed genes were immune system process, response to stimulus, cell activation and skeletal system development, for the overexpressed genes, and unfolded protein binding and stress response, for the underexpressed genes, which included five heat shock proteins. Adipose tissue alterations and impaired stress response are linked to inflammation and, in turn, to adipose tissue secretory activity, similar to what is observed in human obesity. Our results provide the opportunity to identify biomarkers of carcass fat traits to improve the pig production chain and to identify genetic factors that regulate the observed differential expression.
Collapse
Affiliation(s)
- P Zambonelli
- Department of Agricultural and-Food Sciences (DISTAL), Bologna University, Via Fratelli Rosselli 107, 42123, Reggio Emilia, Italy
| | - E Gaffo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy
| | - M Zappaterra
- Department of Agricultural and-Food Sciences (DISTAL), Bologna University, Via Fratelli Rosselli 107, 42123, Reggio Emilia, Italy
| | - S Bortoluzzi
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121, Padova, Italy
| | - R Davoli
- Department of Agricultural and-Food Sciences (DISTAL), Bologna University, Via Fratelli Rosselli 107, 42123, Reggio Emilia, Italy
| |
Collapse
|
28
|
Vieira-Potter VJ. Response to "Perivascular adipose tissue and inflammation. Obesity (Silver Spring) 2016; 24:548. [PMID: 26854016 DOI: 10.1002/oby.21450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 11/12/2022]
|
29
|
Gondret F, Vincent A, Houée-Bigot M, Siegel A, Lagarrigue S, Louveau I, Causeur D. Molecular alterations induced by a high-fat high-fiber diet in porcine adipose tissues: variations according to the anatomical fat location. BMC Genomics 2016; 17:120. [PMID: 26892011 PMCID: PMC4758018 DOI: 10.1186/s12864-016-2438-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/05/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Changing the energy and nutrient source for growing animals may be an effective way of limiting adipose tissue expansion, a response which may depend on the genetic background of the animals. This study aims to describe the transcriptional modulations present in the adipose tissues of two pig lines divergently selected for residual feed intake which were either fed a high-fat high-fiber (HF) diet or an isocaloric low-fat high-starch diet (LF). RESULTS Transcriptomic analysis using a porcine microarray was performed on 48 pigs (n = 12 per diet and per line) in both perirenal (PRAT) and subcutaneous (SCAT) adipose tissues. There was no interaction between diet and line on either adiposity or transcriptional profiles, so that the diet effect was inferred independently of the line. Irrespective of line, the relative weights of the two fat depots were lower in HF pigs than in LF pigs after 58 days on dietary treatment. In the two adipose tissues, the most apparent effect of the HF diet was the down-regulation of several genes associated with the ubiquitin-proteasome system, which therefore may be associated with dietary-induced modulations in genes acting in apoptotic and cell cycle regulatory pathways. Genes involved in glucose metabolic processes were also down-regulated by the HF diet, with no significant variation or decreased expression of important lipid-related genes such as the low-density lipoprotein receptor and leptin in the two fat pads. The master regulators of glucose and fatty acid homeostasis SREBF1 and MLXIPL, and peroxisome proliferator-activated receptor (PPAR)δ and its heterodimeric partner RXRA were down-regulated by the HF diet. PPARγ which has pleiotropic functions including lipid metabolism and adipocyte differentiation, was however up-regulated by this diet in PRAT and SCAT. Dietary-related modulations in the expression of genes associated with immunity and inflammation were mainly revealed in PRAT. CONCLUSION A high-fat high-fiber diet depressed glucose and lipid anabolic molecular pathways, thus counteracting adipose tissue expansion. Interaction effects between dietary intake of fiber and lipids on gene expression may modulate innate immunity and inflammation, a response which is of interest with regard to chronic inflammation and its adverse effects on health and performance.
Collapse
Affiliation(s)
- Florence Gondret
- INRA, UMR1348 Pegase, F-35590, Saint-Gilles, France. .,Agrocampus-Ouest, UMR1348 Pegase, F-35000, Rennes, France.
| | - Annie Vincent
- INRA, UMR1348 Pegase, F-35590, Saint-Gilles, France. .,Agrocampus-Ouest, UMR1348 Pegase, F-35000, Rennes, France.
| | | | - Anne Siegel
- CNRS-Université de Rennes 1-INRIA, UMR6074 IRISA, Campus de Beaulieu, 35042, Rennes, Cedex, France.
| | - Sandrine Lagarrigue
- INRA, UMR1348 Pegase, F-35590, Saint-Gilles, France. .,Agrocampus-Ouest, UMR1348 Pegase, F-35000, Rennes, France.
| | - Isabelle Louveau
- INRA, UMR1348 Pegase, F-35590, Saint-Gilles, France. .,Agrocampus-Ouest, UMR1348 Pegase, F-35000, Rennes, France.
| | - David Causeur
- Agrocampus-Ouest, UMR6625 IRMAR, F-35000, Rennes, France.
| |
Collapse
|
30
|
Rapid Alterations in Perirenal Adipose Tissue Transcriptomic Networks with Cessation of Voluntary Running. PLoS One 2015; 10:e0145229. [PMID: 26678390 PMCID: PMC4683046 DOI: 10.1371/journal.pone.0145229] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/30/2015] [Indexed: 12/20/2022] Open
Abstract
In maturing rats, the growth of abdominal fat is attenuated by voluntary wheel running. After the cessation of running by wheel locking, a rapid increase in adipose tissue growth to a size that is similar to rats that have never run (i.e. catch-up growth) has been previously reported by our lab. In contrast, diet-induced increases in adiposity have a slower onset with relatively delayed transcriptomic responses. The purpose of the present study was to identify molecular pathways associated with the rapid increase in adipose tissue after ending 6 wks of voluntary running at the time of puberty. Age-matched, male Wistar rats were given access to running wheels from 4 to 10 weeks of age. From the 10th to 11th week of age, one group of rats had continued wheel access, while the other group had one week of wheel locking. Perirenal adipose tissue was extracted, RNA sequencing was performed, and bioinformatics analyses were executed using Ingenuity Pathway Analysis (IPA). IPA was chosen to assist in the understanding of complex ‘omics data by integrating data into networks and pathways. Wheel locked rats gained significantly more fat mass and significantly increased body fat percentage between weeks 10–11 despite having decreased food intake, as compared to rats with continued wheel access. IPA identified 646 known transcripts differentially expressed (p < 0.05) between continued wheel access and wheel locking. In wheel locked rats, IPA revealed enrichment of transcripts for the following functions: extracellular matrix, macrophage infiltration, immunity, and pro-inflammatory. These findings suggest that increases in visceral adipose tissue that accompanies the cessation of pubertal physical activity are associated with the alteration of multiple pathways, some of which may potentiate the development of pubertal obesity and obesity-associated systemic low-grade inflammation that occurs later in life.
Collapse
|
31
|
Vieira-Potter VJ, Lee S, Bayless DS, Scroggins RJ, Welly RJ, Fleming NJ, Smith TN, Meers GM, Hill MA, Rector RS, Padilla J. Disconnect between adipose tissue inflammation and cardiometabolic dysfunction in Ossabaw pigs. Obesity (Silver Spring) 2015; 23:2421-9. [PMID: 26524201 PMCID: PMC4701582 DOI: 10.1002/oby.21252] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The Ossabaw pig is emerging as an attractive model of human cardiometabolic disease because of its size and susceptibility to atherosclerosis, among other characteristics. The relationship between adipose tissue inflammation and metabolic dysfunction in this model was investigated here. METHODS Young female Ossabaw pigs were fed a Western-style high-fat diet (HFD) (n = 4) or control low-fat diet (LFD) (n = 4) for a period of 9 months and compared for cardiometabolic outcomes and adipose tissue inflammation. RESULTS The HFD-fed "OBESE" pigs were 2.5 times heavier (P < 0.001) than LFD-fed "LEAN" pigs and developed severe obesity. HFD feeding caused pronounced dyslipidemia, hypertension, and insulin resistance (systemic and adipose), as well as induction of inflammatory genes, impairments in vasomotor reactivity to insulin, and atherosclerosis in the coronary arteries. Remarkably, visceral, subcutaneous, and perivascular adipose tissue inflammation (via FACS analysis and RT-PCR) was not increased in OBESE pigs, nor were circulating inflammatory cytokines. CONCLUSIONS These findings reveal a disconnect between adipose tissue inflammation and cardiometabolic dysfunction induced by Western diet feeding in the Ossabaw pig model.
Collapse
Affiliation(s)
| | - Sewon Lee
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Division of Sport Science & Sport Science Institute, Incheon National University, Incheon, South Korea
| | - David S. Bayless
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | | | - Rebecca J. Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | | | - Thomas N. Smith
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Grace M. Meers
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
| | - R. Scott Rector
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
- Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Child Health, University of Missouri, Columbia, MO
| |
Collapse
|
32
|
Toedebusch RG, Ruegsegger GN, Braselton JF, Heese AJ, Hofheins JC, Childs TE, Thyfault JP, Booth FW. AMPK agonist AICAR delays the initial decline in lifetime-apex V̇o2 peak, while voluntary wheel running fails to delay its initial decline in female rats. Physiol Genomics 2015; 48:101-15. [PMID: 26578698 DOI: 10.1152/physiolgenomics.00078.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022] Open
Abstract
There has never been an outcome measure for human health more important than peak oxygen consumption (V̇o2 peak), yet little is known regarding the molecular triggers for its lifetime decline with aging. We examined the ability of physical activity or 5 wk of 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) administration to delay the initial aging-induced decline in lifetime-apex V̇o2 peak and potential underlying molecular mechanisms. Experiment 1 consisted of female rats with (RUN) and without (NO RUN) running wheels, while experiment 2 consisted of female nonrunning rats getting the AMPK agonist AICAR (0.5 mg/g/day) subcutaneously for 5 wk beginning at 17 wk of age. All rats underwent frequent, weekly or biweekly V̇o2 peak tests beginning at 10 wk of age. In experiment 1, lifetime-apex V̇o2 peak occurred at 19 wk of age in both RUN and NO RUN and decreased thereafter. V̇o2 peak measured across experiment 1 was ∼25% higher in RUN than in NO RUN. In experiment 2, AICAR delayed the chronological age observed in experiment 1 by 1 wk, from 19 wk to 20 wk of age. RUN and NO RUN showed different skeletal muscle transcriptomic profiles both pre- and postapex. Additionally, growth and development pathways are differentially regulated between RUN and NO RUN. Angiomotin mRNA was downregulated postapex in RUN and NO RUN. Furthermore, strong significant correlations to V̇o2 peak and trends for decreased protein concentration supports angiomotin's potential importance in our model. Contrary to our primary hypothesis, wheel running was not sufficient to delay the chronological age of lifetime-apex V̇o2 peak decline, whereas AICAR delayed it 1 wk.
Collapse
Affiliation(s)
- Ryan G Toedebusch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | | | - Joshua F Braselton
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Alexander J Heese
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John C Hofheins
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Tom E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|