1
|
Haymet AB, Lau C, Cho C, O'Loughlin S, Pinto NV, McGiffin DC, Vallely MP, Suen JY, Fraser JF. A Novel Porcine Model of Bilateral Hindlimb Bypass Graft Surgery Integrating Transit Time Flowmetry. J Cardiothorac Surg 2024; 19:661. [PMID: 39702209 DOI: 10.1186/s13019-024-03192-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Bypass graft surgery is a key surgical intervention for ischemic heart disease (coronary bypass graft surgery) and critical limb ischemia (peripheral bypass graft surgery). Graft occlusion remains a significant clinical problem for both types. Further research into the pathobiological mechanisms of graft occlusion are needed in order to design targeted therapeutic strategies. METHODS Three Large White female pigs (mean weight 52.3 +/- 4.4 kg) received general anaesthesia prior to surgery. The external jugular vein was harvested bilaterally, and a bilateral femoral peripheral arterial bypass was performed, with the superficial femoral artery permanently ligated. The grafts were interrogated immediately post operatively on-table using Medistim MiraQ transit time flowmetry system (Medistim, Oslo, Norway) to assess graft performance. On postoperative day three, the pigs were returned to the operating room, and the grafts were interrogated once again using transit time flowmetry. RESULTS Six out of six (100%) successful bilateral EJV to femoral artery bypass grafts were performed. All pigs were successfully recovered, and returned to the operating room at postoperative day 3. The wounds were re-opened and the grafts were inspected. Postoperative graft assessment was performed with transit time flowmetry using the Medistim MiraQTM system (Medistim, Oslo, Norway), demonstrating all grafts were patent (100%). CONCLUSION This model may serve as a platform to gain further mechanistic insight into graft failure pathobiology. By combining a bilateral graft model with gold-standard transit time flowmetry, longitudinal experimentation of targeted therapeutic interventions to combat graft failure may be further studied with improved objectivity.
Collapse
Affiliation(s)
- Andrew B Haymet
- Critical Care Research Group, The Prince Charles Hospital, L1 Clinical Sciences Building, Chermside, QLD, 4032, Australia.
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia.
| | - Cora Lau
- UQ Biological Resources, Herston Medical Research Centre, University of Queensland, Herston, QLD, Australia
| | - Christina Cho
- UQ Biological Resources, Herston Medical Research Centre, University of Queensland, Herston, QLD, Australia
| | - Sean O'Loughlin
- UQ Biological Resources, Herston Medical Research Centre, University of Queensland, Herston, QLD, Australia
| | - Nigel V Pinto
- Department of Vascular Surgery, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - David C McGiffin
- Department of Cardiothoracic Surgery, The Alfred Hospital, Melbourne, VIC, Australia
| | - Michael P Vallely
- Department of Cardiothoracic Surgery, St George Hospital, Kogarah, NSW, Australia
- Macquarie University Hospital, Macquarie Park, NSW, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, L1 Clinical Sciences Building, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, L1 Clinical Sciences Building, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
2
|
Zhu B, Gupta K, Cui K, Wang B, Malovichko MV, Han X, Li K, Wu H, Arulsamy KS, Singh B, Gao J, Wong S, Cowan DB, Wang D, Biddinger S, Srivastava S, Shi J, Chen K, Chen H. Targeting Liver Epsins Ameliorates Dyslipidemia in Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609742. [PMID: 39253478 PMCID: PMC11383288 DOI: 10.1101/2024.08.26.609742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Rationale Low density cholesterol receptor (LDLR) in the liver is critical for the clearance of low-density lipoprotein cholesterol (LDL-C) in the blood. In atherogenic conditions, proprotein convertase subtilisin/kexin 9 (PCSK9) secreted by the liver, in a nonenzymatic fashion, binds to LDLR on the surface of hepatocytes, preventing its recycling and enhancing its degradation in lysosomes, resulting in reduced LDL-C clearance. Our recent studies demonstrate that epsins, a family of ubiquitin-binding endocytic adaptors, are critical regulators of atherogenicity. Given the fundamental contribution of circulating LDL-C to atherosclerosis, we hypothesize that liver epsins promote atherosclerosis by controlling LDLR endocytosis and degradation. Objective We will determine the role of liver epsins in promoting PCSK9-mediated LDLR degradation and hindering LDL-C clearance to propel atherosclerosis. Methods and Results We generated double knockout mice in which both paralogs of epsins, namely, epsin-1 and epsin-2, are specifically deleted in the liver (Liver-DKO) on an ApoE -/- background. We discovered that western diet (WD)-induced atherogenesis was greatly inhibited, along with diminished blood cholesterol and triglyceride levels. Mechanistically, using scRNA-seq analysis on cells isolated from the livers of ApoE-/- and ApoE-/- /Liver-DKO mice on WD, we found lipogenic Alb hi hepatocytes to glycogenic HNF4α hi hepatocytes transition in ApoE-/- /Liver-DKO. Subsequently, gene ontology analysis of hepatocyte-derived data revealed elevated pathways involved in LDL particle clearance and very-low-density lipoprotein (VLDL) particle clearance under WD treatment in ApoE-/- /Liver-DKO, which was coupled with diminished plasma LDL-C levels. Further analysis using the MEBOCOST algorithm revealed enhanced communication score between LDLR and cholesterol, suggesting elevated LDL-C clearance in the ApoE-/- Liver-DKO mice. In addition, we showed that loss of epsins in the liver upregulates of LDLR protein level. We further showed that epsins bind LDLR via the ubiquitin-interacting motif (UIM), and PCSK9-triggered LDLR degradation was abolished by depletion of epsins, preventing atheroma progression. Finally, our therapeutic strategy, which involved targeting liver epsins with nanoparticle-encapsulated siRNAs, was highly efficacious at inhibiting dyslipidemia and impeding atherosclerosis. Conclusions Liver epsins promote atherogenesis by mediating PCSK9-triggered degradation of LDLR, thus raising the circulating LDL-C levels. Targeting epsins in the liver may serve as a novel therapeutic strategy to treat atherosclerosis by suppression of PCSK9-mediated LDLR degradation.
Collapse
Affiliation(s)
- Bo Zhu
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Krishan Gupta
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kui Cui
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Beibei Wang
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Marina V Malovichko
- Department of Medicine, Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, United States
| | - Xiangfei Han
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathryn Li
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Hao Wu
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Kulandai Samy Arulsamy
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Bandana Singh
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Jianing Gao
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Scott Wong
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Douglas B Cowan
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Dazhi Wang
- College of Medicine Molecular Pharmacology, University of South Florida, Tampa, FL, United States
| | - Sudha Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sanjay Srivastava
- Department of Medicine, Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, United States
| | - Jinjun Shi
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kaifu Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Jia Y, Zhao Y, Niu M, Zhao C, Li X, Chen H. Preliminary study of metabonomic changes during the progression of atherosclerosis in miniature pigs. Animal Model Exp Med 2024; 7:419-432. [PMID: 38923366 PMCID: PMC11369038 DOI: 10.1002/ame2.12462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND To explore potential biomarkers for early diagnosis of atherosclerosis (AS) and provide basic data for further research on AS, the characteristics of serum metabolomics during the progression of AS in mini-pigs were observed dynamically. METHODS An AS model in Bama miniature pigs was established by a high-cholesterol and high-fat diet. Fasting serum samples were collected monthly for metabolomics and serum lipid detection. At the end of the treatment period, pathological analysis of the abdominal aorta and coronary artery was performed to evaluate the lesions of AS, thereby distinguishing the susceptibility of mini-pigs to AS. The metabolomics was detected using a high-resolution untargeted metabolomic approach. Statistical analysis was used to identify metabolites associated with AS susceptibility. RESULTS Based on pathological analysis, mini-pigs were divided into two groups: a susceptible group (n = 3) and a non-susceptible group (n = 6). A total of 1318 metabolites were identified, with significant shifting of metabolic profiles over time in both groups. Dynamic monitoring analysis highlighted 57 metabolites that exhibited an obvious trend of differential changes between two groups with the advance of time. The KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis indicated significant disorders in cholesterol metabolism, primary bile acid metabolism, histidine metabolism, as well as taurine and hypotaurine metabolism. CONCLUSIONS During the progression of AS in mini-pigs induced by high-cholesterol/high-fat diet, the alterations in serum metabolic profile exhibited a time-dependent pattern, accompanied by notable disturbances in lipid metabolism, cholesterol metabolism, and amino acid metabolism. These metabolites may become potential biomarkers for early diagnosis of AS.
Collapse
Affiliation(s)
- Yunxiao Jia
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Yuqiong Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Miaomiao Niu
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Changqi Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Xuezhuang Li
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Hua Chen
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| |
Collapse
|
4
|
Ding D, Zhao Y, Jia Y, Niu M, Li X, Zheng X, Chen H. Identification of novel genes associated with atherosclerosis in Bama miniature pig. Animal Model Exp Med 2024; 7:377-387. [PMID: 38720469 PMCID: PMC11228093 DOI: 10.1002/ame2.12412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/20/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic cardiovascular disease of great concern. However, it is difficult to establish a direct connection between conventional small animal models and clinical practice. The pig's genome, physiology, and anatomy reflect human biology better than other laboratory animals, which is crucial for studying the pathogenesis of atherosclerosis. METHODS We used whole-genome sequencing data from nine Bama minipigs to perform a genome-wide linkage analysis, and further used bioinformatic tools to filter and identify underlying candidate genes. Candidate gene function prediction was performed using the online prediction tool STRING 12.0. Immunohistochemistry and immunofluorescence were used to detect the expression of proteins encoded by candidate genes. RESULTS We mapped differential single nucleotide polymorphisms (SNPs) to genes and obtained a total of 102 differential genes, then we used GO and KEGG pathway enrichment analysis to identify four candidate genes, including SLA-1, SLA-2, SLA-3, and TAP2. nsSNPs cause changes in the primary and tertiary structures of SLA-I and TAP2 proteins, the primary structures of these two proteins have undergone amino acid changes, and the tertiary structures also show slight changes. In addition, immunohistochemistry and immunofluorescence results showed that the expression changes of TAP2 protein in coronary arteries showed a trend of increasing from the middle layer to the inner layer. CONCLUSIONS We have identified SLA-I and TAP2 as potential susceptibility genes of atherosclerosis, highlighting the importance of antigen processing and immune response in atherogenesis.
Collapse
Affiliation(s)
- Dengfeng Ding
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Yuqiong Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Yunxiao Jia
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Miaomiao Niu
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Xuezhuang Li
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Xinou Zheng
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Hua Chen
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
5
|
Sams A, Haanes KA, Holm A, Kazantzi S, Mikkelsen LF, Edvinsson L, Brain S, Sheykhzade M. Heterogeneous vasomotor responses in segments from Göttingen Minipigs coronary, cerebral, and mesenteric artery: A comparative study. Vascul Pharmacol 2023; 153:107231. [PMID: 37730143 DOI: 10.1016/j.vph.2023.107231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
Göttingen Minipigs (GM) are used as an important preclinical model for cardiovascular safety pharmacology and for evaluation of cardiovascular drug targets. To improve the translational value of the GM model, the current study represents a basic characterization of vascular responses to endothelial regulators and sympathetic, parasympathetic, and sensory neurotransmitters in different anatomical origins. The aim of the current comparative and descriptive study is to use myography to characterize the vasomotor responses of coronary artery isolated from GM and compare the responses to those obtained from parallel studies using cerebral and mesenteric arteries. The selected agonists for sympathetic (norepinephrine), parasympathetic (carbachol), sensory (calcitonin gene-related peptide, CGRP), and endothelial pathways (endothelin-1, ET-1, and bradykinin) were used for comparison. Further, the robust nature of the vasomotor responses was evaluated after 24 h of cold storage of vascular tissue mimicking the situation under which human biopsies are often kept before experiments or grafting is feasible. Results show that bradykinin and CGRP consistently dilated, and endothelin consistently contracted artery segments from coronary, cerebral, and mesenteric origin. By comparison, norepinephrine and carbachol, had responses that varied with the anatomical source of the tissues. To support the basic characterization of GM vasomotor responses, we demonstrated the presence of mRNA encoding selected vascular receptors (CGRP- and ETA-receptors) in fresh artery segments. In conclusion, the vasomotor responses of isolated coronary, cerebral, and mesenteric arteries to selected agonists of endothelial, sympathetic, parasympathetic, and sensory pathways are different and the phenotypes are similar to sporadic human findings.
Collapse
Affiliation(s)
- Anette Sams
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark; Epoqe Pharma, Ole Maaloes Vej 3, 2200 Copenhagen N, Denmark.
| | | | - Anja Holm
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark; Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Spyridoula Kazantzi
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
| | | | - Lars Edvinsson
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
| | - Susan Brain
- Section of Vascular Biology & Inflammation, School of Cardiovascular Medicine & Research, BHF Centre of Excellence, King's College London, London, United Kingdom
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
6
|
Cheng J, Huang H, Chen Y, Wu R. Nanomedicine for Diagnosis and Treatment of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304294. [PMID: 37897322 PMCID: PMC10754137 DOI: 10.1002/advs.202304294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/11/2023] [Indexed: 10/30/2023]
Abstract
With the changing disease spectrum, atherosclerosis has become increasingly prevalent worldwide and the associated diseases have emerged as the leading cause of death. Due to their fascinating physical, chemical, and biological characteristics, nanomaterials are regarded as a promising tool to tackle enormous challenges in medicine. The emerging discipline of nanomedicine has filled a huge application gap in the atherosclerotic field, ushering a new generation of diagnosis and treatment strategies. Herein, based on the essential pathogenic contributors of atherogenesis, as well as the distinct composition/structural characteristics, synthesis strategies, and surface design of nanoplatforms, the three major application branches (nanodiagnosis, nanotherapy, and nanotheranostic) of nanomedicine in atherosclerosis are elaborated. Then, state-of-art studies containing a sequence of representative and significant achievements are summarized in detail with an emphasis on the intrinsic interaction/relationship between nanomedicines and atherosclerosis. Particularly, attention is paid to the biosafety of nanomedicines, which aims to pave the way for future clinical translation of this burgeoning field. Finally, this comprehensive review is concluded by proposing unresolved key scientific issues and sharing the vision and expectation for the future, fully elucidating the closed loop from atherogenesis to the application paradigm of nanomedicines for advancing the early achievement of clinical applications.
Collapse
Affiliation(s)
- Jingyun Cheng
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Hui Huang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Rong Wu
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
7
|
Zhao Y, Niu M, Jia Y, Yuan J, Xiang L, Dai X, Wang G, Chen H. Establishment of type 2 diabetes mellitus models using streptozotocin after 3 months high-fat diet in Bama minipigs. Anim Biotechnol 2023; 34:2295-2312. [PMID: 35749713 DOI: 10.1080/10495398.2022.2088548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
In the past twenty years, the number of adults with diabetes has tripled. Most studies have been conducted using rodent models of type 2 diabetes mellitus (T2DM), and the developed drugs have low clinical conversion efficiency. Therefore, it is urgent to establish a more human-like large animal model to explore T2DM pathogenesis and formulate new disease prevention and control strategies. This study was designed to establish and validate a T2DM model using minipigs fed a high-fat or high-cholesterol/high-fat diet and injected with low-dose streptozotocin (STZ). We examined the influence of the STZ injection timing with a diet high in fat (HFD) compared with one high in cholesterol and fat (HCFD) on the atherosclerotic lesions accelerated by T2DM. Male Bama minipigs (n = 24) were randomly divided into five groups. The control group was fed a normal diet for 9 months. The STZ + HFD and STZ + HCFD groups were infused with 90 mg/kg STZ and then fed a high-fat diet or high-cholesterol and high-fat diet for 9 months, respectively. The HFD + STZ and HCFD + STZ groups were fed a high-fat diet or a high-cholesterol and high-fat diet, respectively, for 9 months (after 3 months, these pigs were injected intravenously with 90 mg/kg STZ). During the induction period, animal body weight, BMI, and serum GLU, INS, TG, TC, HDL-C, LDL-C, FFA, ALT, AST, CRE, and BUN were detected monthly intervals. IVGTT and insulin release tests were performed at 3-month intervals. At the end of the test, the coronary artery and abdominal aorta were examined by computed tomography and pathological observations, and the thickness of the basement membrane of the capillary of the retina and kidney glomerulus was measured under a transmission electron microscope. The serum glucose concentrations were normal in all groups except the HFD + STZ and HCFD + STZ groups. Animals fed an HFD for 9 months did not develop apparent atherosclerotic lesions, but atherosclerotic lesions were seen in the animals fed an HCFD. Hyperglycemia accelerated the formation of atherosclerotic lesions on the intimal surface of the abdominal aorta. Low-dose STZ after 3 months of HFD or HCFD successfully established a T2DM model in minipigs. The HFD did not induce apparent atherosclerotic lesions, but these were seen with the HCFD. Hyperglycemia accelerated atherosclerosis in the minipigs.
Collapse
Affiliation(s)
- Yuqiong Zhao
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Miaomiao Niu
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Yunxiao Jia
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Jifang Yuan
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Lei Xiang
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Xin Dai
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| | - Guisheng Wang
- Radiology Department of No. 3 Clinical Center, Chinese PLA General Hospital, Beijing, China
| | - Hua Chen
- Laboratory Animal Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Sukhanov S, Higashi Y, Yoshida T, Danchuk S, Alfortish M, Goodchild T, Scarborough A, Sharp T, Jenkins JS, Garcia D, Ivey J, Tharp DL, Schumacher J, Rozenbaum Z, Kolls JK, Bowles D, Lefer D, Delafontaine P. Insulin-like growth factor 1 reduces coronary atherosclerosis in pigs with familial hypercholesterolemia. JCI Insight 2023; 8:e165713. [PMID: 36602878 PMCID: PMC9990768 DOI: 10.1172/jci.insight.165713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Although murine models of coronary atherosclerotic disease have been used extensively to determine mechanisms, limited new therapeutic options have emerged. Pigs with familial hypercholesterolemia (FH pigs) develop complex coronary atheromas that are almost identical to human lesions. We reported previously that insulin-like growth factor 1 (IGF-1) reduced aortic atherosclerosis and promoted features of stable plaque in a murine model. We administered human recombinant IGF-1 or saline (control) in atherosclerotic FH pigs for 6 months. IGF-1 decreased relative coronary atheroma in vivo (intravascular ultrasound) and reduced lesion cross-sectional area (postmortem histology). IGF-1 increased plaque's fibrous cap thickness, and reduced necrotic core, macrophage content, and cell apoptosis, consistent with promotion of a stable plaque phenotype. IGF-1 reduced circulating triglycerides, markers of systemic oxidative stress, and CXCL12 chemokine levels. We used spatial transcriptomics (ST) to identify global transcriptome changes in advanced plaque compartments and to obtain mechanistic insights into IGF-1 effects. ST analysis showed that IGF-1 suppressed FOS/FOSB factors and gene expression of MMP9 and CXCL14 in plaque macrophages, suggesting possible involvement of these molecules in IGF-1's effect on atherosclerosis. Thus, IGF-1 reduced coronary plaque burden and promoted features of stable plaque in a pig model, providing support for consideration of clinical trials.
Collapse
Affiliation(s)
- Sergiy Sukhanov
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yusuke Higashi
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tadashi Yoshida
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Svitlana Danchuk
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Mitzi Alfortish
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Traci Goodchild
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Amy Scarborough
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Thomas Sharp
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | | | | - Jan Ivey
- Ochsner Medical Center, New Orleans, Louisiana, USA
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - Jeffrey Schumacher
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Zach Rozenbaum
- Tulane University School of Medicine, New Orleans, Louisiana, USA
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jay K. Kolls
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Douglas Bowles
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - David Lefer
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | |
Collapse
|
9
|
van der Velden J, Asselbergs FW, Bakkers J, Batkai S, Bertrand L, Bezzina CR, Bot I, Brundel BJJM, Carrier L, Chamuleau S, Ciccarelli M, Dawson D, Davidson SM, Dendorfer A, Duncker DJ, Eschenhagen T, Fabritz L, Falcão-Pires I, Ferdinandy P, Giacca M, Girao H, Gollmann-Tepeköylü C, Gyongyosi M, Guzik TJ, Hamdani N, Heymans S, Hilfiker A, Hilfiker-Kleiner D, Hoekstra AG, Hulot JS, Kuster DWD, van Laake LW, Lecour S, Leiner T, Linke WA, Lumens J, Lutgens E, Madonna R, Maegdefessel L, Mayr M, van der Meer P, Passier R, Perbellini F, Perrino C, Pesce M, Priori S, Remme CA, Rosenhahn B, Schotten U, Schulz R, Sipido KR, Sluijter JPG, van Steenbeek F, Steffens S, Terracciano CM, Tocchetti CG, Vlasman P, Yeung KK, Zacchigna S, Zwaagman D, Thum T. Animal models and animal-free innovations for cardiovascular research: current status and routes to be explored. Consensus document of the ESC Working Group on Myocardial Function and the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2022; 118:3016-3051. [PMID: 34999816 PMCID: PMC9732557 DOI: 10.1093/cvr/cvab370] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases represent a major cause of morbidity and mortality, necessitating research to improve diagnostics, and to discover and test novel preventive and curative therapies, all of which warrant experimental models that recapitulate human disease. The translation of basic science results to clinical practice is a challenging task, in particular for complex conditions such as cardiovascular diseases, which often result from multiple risk factors and comorbidities. This difficulty might lead some individuals to question the value of animal research, citing the translational 'valley of death', which largely reflects the fact that studies in rodents are difficult to translate to humans. This is also influenced by the fact that new, human-derived in vitro models can recapitulate aspects of disease processes. However, it would be a mistake to think that animal models do not represent a vital step in the translational pathway as they do provide important pathophysiological insights into disease mechanisms particularly on an organ and systemic level. While stem cell-derived human models have the potential to become key in testing toxicity and effectiveness of new drugs, we need to be realistic, and carefully validate all new human-like disease models. In this position paper, we highlight recent advances in trying to reduce the number of animals for cardiovascular research ranging from stem cell-derived models to in situ modelling of heart properties, bioinformatic models based on large datasets, and state-of-the-art animal models, which show clinically relevant characteristics observed in patients with a cardiovascular disease. We aim to provide a guide to help researchers in their experimental design to translate bench findings to clinical routine taking the replacement, reduction, and refinement (3R) as a guiding concept.
Collapse
Grants
- R01 HL150359 NHLBI NIH HHS
- RG/16/14/32397 British Heart Foundation
- FS/18/37/33642 British Heart Foundation
- PG/17/64/33205 British Heart Foundation
- PG/15/88/31780 British Heart Foundation
- FS/RTF/20/30009, NH/19/1/34595, PG/18/35/33786, CS/17/4/32960, PG/15/88/31780, and PG/17/64/33205 British Heart Foundation
- NC/T001488/1 National Centre for the Replacement, Refinement and Reduction of Animals in Research
- PG/18/44/33790 British Heart Foundation
- CH/16/3/32406 British Heart Foundation
- FS/RTF/20/30009 British Heart Foundation
- NWO-ZonMW
- ZonMW and Heart Foundation for the translational research program
- Dutch Cardiovascular Alliance (DCVA)
- Leducq Foundation
- Dutch Research Council
- Association of Collaborating Health Foundations (SGF)
- UCL Hospitals NIHR Biomedical Research Centre, and the DCVA
- Netherlands CardioVascular Research Initiative CVON
- Stichting Hartekind and the Dutch Research Counsel (NWO) (OCENW.GROOT.2019.029)
- National Fund for Scientific Research, Belgium and Action de Recherche Concertée de la Communauté Wallonie-Bruxelles, Belgium
- Netherlands CardioVascular Research Initiative CVON (PREDICT2 and CONCOR-genes projects), the Leducq Foundation
- ERA PerMed (PROCEED study)
- Netherlands Cardiovascular Research Initiative
- Dutch Heart Foundation
- German Centre of Cardiovascular Research (DZHH)
- Chest Heart and Stroke Scotland
- Tenovus Scotland
- Friends of Anchor and Grampian NHS-Endowments
- National Institute for Health Research University College London Hospitals Biomedical Research Centre
- German Centre for Cardiovascular Research
- European Research Council (ERC-AG IndivuHeart), the Deutsche Forschungsgemeinschaft
- European Union Horizon 2020 (REANIMA and TRAINHEART)
- German Ministry of Education and Research (BMBF)
- Centre for Cardiovascular Research (DZHK)
- European Union Horizon 2020
- DFG
- National Research, Development and Innovation Office of Hungary
- Research Excellence Program—TKP; National Heart Program
- Austrian Science Fund
- European Union Commission’s Seventh Framework programme
- CVON2016-Early HFPEF
- CVON She-PREDICTS
- CVON Arena-PRIME
- European Union’s Horizon 2020 research and innovation programme
- Deutsche Forschungsgemeinschaft
- Volkswagenstiftung
- French National Research Agency
- ERA-Net-CVD
- Fédération Française de Cardiologie, the Fondation pour la Recherche Médicale
- French PIA Project
- University Research Federation against heart failure
- Netherlands Heart Foundation
- Dekker Senior Clinical Scientist
- Health Holland TKI-LSH
- TUe/UMCU/UU Alliance Fund
- south African National Foundation
- Cancer Association of South Africa and Winetech
- Netherlands Heart Foundation/Applied & Engineering Sciences
- Dutch Technology Foundation
- Pie Medical Imaging
- Netherlands Organisation for Scientific Research
- Dr. Dekker Program
- Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation
- Dutch Federation of University Medical Centres
- Netherlands Organization for Health Research and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project
- Netherlands Organization for Scientific Research (NWO) (VICI grant); the European Research Council
- Incyte s.r.l. and from Ministero dell’Istruzione, Università e Ricerca Scientifica
- German Center for Cardiovascular Research (Junior Research Group & Translational Research Project), the European Research Council (ERC Starting Grant NORVAS),
- Swedish Heart-Lung-Foundation
- Swedish Research Council
- National Institutes of Health
- Bavarian State Ministry of Health and Care through the research project DigiMed Bayern
- ERC
- ERA-CVD
- Dutch Heart Foundation, ZonMw
- the NWO Gravitation project
- Ministero dell'Istruzione, Università e Ricerca Scientifica
- Regione Lombardia
- Netherlands Organisation for Health Research and Development
- ITN Network Personalize AF: Personalized Therapies for Atrial Fibrillation: a translational network
- MAESTRIA: Machine Learning Artificial Intelligence Early Detection Stroke Atrial Fibrillation
- REPAIR: Restoring cardiac mechanical function by polymeric artificial muscular tissue
- Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)
- European Union H2020 program to the project TECHNOBEAT
- EVICARE
- BRAV3
- ZonMw
- German Centre for Cardiovascular Research (DZHK)
- British Heart Foundation Centre for Cardiac Regeneration
- British Heart Foundation studentship
- NC3Rs
- Interreg ITA-AUS project InCARDIO
- Italian Association for Cancer Research
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, UK
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sandor Batkai
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Luc Bertrand
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Connie R Bezzina
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Ilze Bot
- Heart Center, Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Steven Chamuleau
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Odontology, University of Salerno, Fisciano (SA), Italy
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- University Center of Cardiovascular Sciences and Department of Cardiology, University Heart Center Hamburg, Germany and Institute of Cardiovascular Sciences, University of Birmingham, UK
| | - Ines Falcão-Pires
- UnIC - Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Portugal
| | - Péter Ferdinandy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Henrique Girao
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, Faculty of Medicine, Coimbra, Portugal
- Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | | | - Mariann Gyongyosi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Tomasz J Guzik
- Instutute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Nazha Hamdani
- Division Cardiology, Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Andres Hilfiker
- Department for Cardiothoracic, Transplant, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Comprehensive Cancer Centre, Philipps-Universität Marburg, Germany
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Sébastien Hulot
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Linda W van Laake
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Tim Leiner
- Department of Radiology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27B, 48149 Muenster, Germany
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, 56124 Pisa, Italy
- Department of Internal Medicine, Cardiology Division, University of Texas Medical School in Houston, Houston, TX, USA
| | - Lars Maegdefessel
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500AE Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Filippo Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro cardiologico Monzino, IRCCS, Milan, Italy
| | - Silvia Priori
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, Pavia, Italy
- University of Pavia, Pavia, Italy
| | - Carol Ann Remme
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Bodo Rosenhahn
- Institute for information Processing, Leibniz University of Hanover, 30167 Hannover, Germany
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karin R Sipido
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, Regenerative Medicine Center Utrecht, Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Steenbeek
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center for Clinical and Translational Research (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Patricia Vlasman
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Amsterdam UMC, Vrije Universiteit, Surgery, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Dayenne Zwaagman
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Thomas Thum
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
10
|
Sturek M. Introduction to ion transport and membrane interactions in vascular health and disease. CURRENT TOPICS IN MEMBRANES 2022; 90:1-11. [PMID: 36368870 DOI: 10.1016/bs.ctm.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cardiovascular disease is on the rise, partially due to the continued increase in metabolic syndrome. Advances in basic research on vascular ion transport have the potential to provide targets for therapeutic interventions. Vascular specificity, which includes different vascular beds having different characteristics and the macro- vs. microvasculature, is a vitally important variable in characterization of ion transport. At the cellular level, targeted fluorescent biosensors for Ca2+, super-resolution microscopy, and organelle patch clamp electrophysiology enable more detailed studies. The "MetS/diabetes milieu" includes increased and decreased insulin, and increased glucose, increased LDL/HDL cholesterol and triglycerides, and increased blood pressure. The duration and severity of MetS/diabetes components certainly affect the vascular phenotype and ion transport and membrane interactions. A combination of in vivo animal models and in vitro cell models to study ion transport in MetS/diabetes conditions is optimal. Gene editing and selective pharmacological tools should be used after or in conjunction with characterization of ion transport in vascular health and disease phenotypes. This is critical to determining the causal role of Ca2+ signaling in modulation of vascular phenotype. The ion transport and membrane interactions that are measured are typically only a snapshot in time in these dynamic processes occurring over the progression of health and disease. It is imperative that this concept be considered in the planning of long-term studies of vascular disease, ion transport experiments, and interpretation of the data. Future directions for our contributors' research will advance the field.
Collapse
Affiliation(s)
- Michael Sturek
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
11
|
Wahl L, Raschke M, Wittmann J, Regler A, Heelemann S, Brandsch C, Stangl GI, Vervuert I. Effects of atherogenic diet supplemented with fermentable carbohydrates on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. PLoS One 2022; 17:e0275214. [PMID: 36206259 PMCID: PMC9543622 DOI: 10.1371/journal.pone.0275214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Fermentable carbohydrates are gaining interest in the field of human nutrition because of their benefits in obesity-related comorbidities. The aim of this study was to investigate the influence of fermentable carbohydrates, such as pectin and inulin, in an atherogenic diet on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. Forty-eight healthy pigs aged five months were divided into four feeding groups (n = 10) and one baseline group (n = 8). Three feeding groups received an atherogenic diet (38% crisps, 10% palm fat, and 2% sugar with or without supplementation of 5% pectin or inulin), and one group received a conventional diet over 15 weeks. Feed intake, weight gain, body condition score, and back fat thickness were monitored regularly. Blood and fecal samples were collected monthly to assess the metabolites associated with high cardiovascular risk and fat content, respectively. At the end of 15 weeks, the coronary arteries of the pigs were analyzed for atherosclerotic plaque formation. Independent of supplementation, significant changes were observed in lipid metabolism, such as an increase in triglycerides, bile acids, and cholesterol in serum, in all groups fed atherogenic diets in comparison to the conventional group. Serum metabolome analysis showed differentiation of the feeding groups by diet (atherogenic versus conventional diet) but not by supplementation with pectin or inulin. Cardiovascular lesions were found in all feeding groups and in the baseline group. Supplementation of pectin or inulin in the atherogenic diet had no significant impact on cardiovascular lesion size. Saddleback pigs can develop naturally occurring plaques in coronary arteries. Therefore, this pig model offers potential for further research on the effects of dietary intervention on obesity-related comorbidities, such as cardiovascular lesions, in humans.
Collapse
Affiliation(s)
- Lisa Wahl
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Melina Raschke
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | | | | | - Corinna Brandsch
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriele I. Stangl
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ingrid Vervuert
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,* E-mail:
| |
Collapse
|
12
|
Cluzel GL, Ryan PM, Herisson FM, Caplice NM. High-fidelity porcine models of metabolic syndrome: a contemporary synthesis. Am J Physiol Endocrinol Metab 2022; 322:E366-E381. [PMID: 35224983 DOI: 10.1152/ajpendo.00413.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review aims to describe and compare porcine models of metabolic syndrome. This syndrome and its associated secondary comorbidities are set to become the greatest challenge to healthcare providers and policy makers in the coming century. However, an incomplete understanding of the pathogenesis has left significant knowledge gaps in terms of efficacious therapeutics. To further our comprehension and, in turn, management of metabolic syndrome, appropriate high-fidelity models of the disease complex are of great importance. In this context, our review aims to assess the most promising porcine models of metabolic syndrome currently available for their similarity to the human phenotype. In addition, we aim to highlight the strengths and shortcomings of each model in an attempt to identify the most appropriate application of each. Although no porcine model perfectly recapitulates the human metabolic syndrome, several pose satisfactory approximations. The Ossabaw miniature swine in particular represents a highly translatable model that develops each of the core parameters of the syndrome with many of the associated secondary comorbidities. Future high-fidelity porcine models of metabolic syndrome need to focus on secondary sequelae replication, which may require extended induction period to reveal.
Collapse
Affiliation(s)
- Gaston L Cluzel
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul M Ryan
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Florence M Herisson
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Noel M Caplice
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Hooshdaran B, Pressly BB, Alferiev IS, Smith JD, Zoltick PW, Tschabrunn CM, Wilensky RL, Gorman RC, Levy RJ, Fishbein I. Stent-based delivery of AAV2 vectors encoding oxidation-resistant apoA1. Sci Rep 2022; 12:5464. [PMID: 35361857 PMCID: PMC8971450 DOI: 10.1038/s41598-022-09524-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022] Open
Abstract
In-stent restenosis (ISR) complicates revascularization in the coronary and peripheral arteries. Apolipoprotein A1 (apoA1), the principal protein component of HDL possesses inherent anti-atherosclerotic and anti-restenotic properties. These beneficial traits are lost when wild type apoA1(WT) is subjected to oxidative modifications. We investigated whether local delivery of adeno-associated viral (AAV) vectors expressing oxidation-resistant apoA1(4WF) preserves apoA1 functionality. The efflux of 3H-cholesterol from macrophages to the media conditioned by endogenously produced apoA1(4WF) was 2.1-fold higher than for apoA1(WT) conditioned media in the presence of hypochlorous acid emulating conditions of oxidative stress. The proliferation of apoA1(WT)- and apoA1(4FW)-transduced rat aortic smooth muscle cells (SMC) was inhibited by 66% ± 10% and 65% ± 11%, respectively, in comparison with non-transduced SMC (p < 0.001). Conversely, the proliferation of apoA1(4FW)-transduced, but not apoA1(WT)-transduced rat blood outgrowth endothelial cells (BOEC) was increased 41% ± 5% (p < 0.001). Both apoA1 transduction conditions similarly inhibited basal and TNFα-induced reactive oxygen species in rat aortic endothelial cells (RAEC) and resulted in the reduced rat monocyte attachment to the TNFα-activated endothelium. AAV2-eGFP vectors immobilized reversibly on stainless steel mesh surfaces through the protein G/anti-AAV2 antibody coupling, efficiently transduced cells in culture modeling stent-based delivery. In vivo studies in normal pigs, deploying AAV2 gene delivery stents (GDS) preloaded with AAV2-eGFP in the coronary arteries demonstrated transduction of the stented arteries. However, implantation of GDS formulated with AAV2-apoA1(4WF) failed to prevent in-stent restenosis in the atherosclerotic vasculature of hypercholesterolemic diabetic pigs. It is concluded that stent delivery of AAV2-4WF while feasible, is not effective for mitigation of restenosis in the presence of severe atherosclerotic disease.
Collapse
Affiliation(s)
- Bahman Hooshdaran
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Benjamin B Pressly
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Ivan S Alferiev
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Jonathan D Smith
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, USA
| | - Philip W Zoltick
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
| | - Cory M Tschabrunn
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert L Wilensky
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert C Gorman
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Robert J Levy
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Ilia Fishbein
- Division of Cardiology, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, CHOP, ARC, Room 702 C, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| |
Collapse
|
14
|
Pig and Mouse Models of Hyperlipidemia and Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:379-411. [PMID: 35237978 DOI: 10.1007/978-1-0716-1924-7_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is a chronic inflammatory disorder that is the underlying cause of most cardiovascular disease. Resident cells of the artery wall and cells of the immune system participate in atherogenesis. This process is influenced by plasma lipoproteins, genetics, and the hemodynamics of the blood flow in the artery. A variety of animal models have been used to study the pathophysiology and mechanisms that contribute to atherosclerotic lesion formation. No model is ideal as each has its own advantages and limitations with respect to manipulation of the atherogenic process and modeling human atherosclerosis and lipoprotein profile. In this chapter we will discuss pig and mouse models of experimental atherosclerosis. The similarity of pig lipoprotein metabolism and the pathophysiology of the lesions in these animals with that of humans is a major advantage. While a few genetically engineered pig models have been generated, the ease of genetic manipulation in mice and the relatively short time frame for the development of atherosclerosis has made them the most extensively used model. Newer approaches to induce hypercholesterolemia in mice have been developed that do not require germline modifications. These approaches will facilitate studies on atherogenic mechanisms.
Collapse
|
15
|
Skydsgaard M, Dincer Z, Haschek WM, Helke K, Jacob B, Jacobsen B, Jeppesen G, Kato A, Kawaguchi H, McKeag S, Nelson K, Rittinghausen S, Schaudien D, Vemireddi V, Wojcinski ZW. International Harmonization of Nomenclature and Diagnostic Criteria (INHAND): Nonproliferative and Proliferative Lesions of the Minipig. Toxicol Pathol 2021; 49:110-228. [PMID: 33393872 DOI: 10.1177/0192623320975373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions) Project (www.toxpath.org/inhand.asp) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in most tissues and organs from the minipig used in nonclinical safety studies. Some of the lesions are illustrated by color photomicrographs. The standardized nomenclature presented in this document is also available electronically on the internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature for lesions in laboratory animals will provide a common language among regulatory and scientific research organizations in different countries and increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
| | - Zuhal Dincer
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | - Wanda M Haschek
- Department of Pathobiology, University of Illinois, Urbana, IL, USA
| | - Kris Helke
- Medical University of South Carolina, Charleston, SC, USA
| | | | - Bjoern Jacobsen
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Gitte Jeppesen
- Charles River Laboratories Copenhagen, Lille Skensved, Denmark
| | - Atsuhiko Kato
- Chugai Pharmaceutical Co, Ltd Research Division, Shizuoka, Japan
| | | | - Sean McKeag
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | | | - Susanne Rittinghausen
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | | | | |
Collapse
|
16
|
Richter C, Hinkel R. Research('s) Sweet Hearts: Experimental Biomedical Models of Diabetic Cardiomyopathy. Front Cardiovasc Med 2021; 8:703355. [PMID: 34368257 PMCID: PMC8342758 DOI: 10.3389/fcvm.2021.703355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Diabetes and the often accompanying cardiovascular diseases including cardiomyopathy represent a complex disease, that is reluctant to reveal the molecular mechanisms and underlying cellular responses. Current research projects on diabetic cardiomyopathy are predominantly based on animal models, in which there are not only obvious advantages, such as genetics that can be traced over generations and the directly measurable influence of dietary types, but also not despisable disadvantages. Thus, many studies are built up on transgenic rodent models, which are partly comparable to symptoms in humans due to their genetic alterations, but on the other hand are also under discussion regarding their clinical relevance in the translation of biomedical therapeutic approaches. Furthermore, a focus on transgenic rodent models ignores spontaneously occurring diabetes in larger mammals (such as dogs or pigs), which represent with their anatomical similarity to humans regarding their cardiovascular situation appealing models for testing translational approaches. With this in mind, we aim to shed light on the currently most popular animal models for diabetic cardiomyopathy and, by weighing the advantages and disadvantages, provide decision support for future animal experimental work in the field, hence advancing the biomedical translation of promising approaches into clinical application.
Collapse
Affiliation(s)
- Claudia Richter
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany.,Partnersite Goettingen, German Center for Cardiovascular Research (DZHK e.V.), Goettingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany.,Partnersite Goettingen, German Center for Cardiovascular Research (DZHK e.V.), Goettingen, Germany.,Stiftung Tierärztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
| |
Collapse
|
17
|
Kim M, Kim HB, Park DS, Cho KH, Hyun DY, Kee HJ, Hong YJ, Jeong MH. A model of atherosclerosis using nicotine with balloon overdilation in a porcine. Sci Rep 2021; 11:13695. [PMID: 34211091 PMCID: PMC8249376 DOI: 10.1038/s41598-021-93229-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/21/2021] [Indexed: 12/31/2022] Open
Abstract
Pigs are important experimental animals for cardiovascular research. Few porcine coronary atherosclerosis models have been developed; however, their induction requires more than six months. We developed a porcine coronary artery atherosclerosis model using nicotine injection with a balloon overdilation. A coronary balloon was placed in the porcine coronary artery and overdilated to induce a mechanical injury. Nicotine was administrated via intramuscular injection every day, and changes in the coronary artery were observed after four weeks. Coronary angiography revealed nicotine injection with a balloon overdilation group showed narrowing of the coronary artery at the injury site. The combination of balloon and nicotine significantly increased the intimal hyperplasia in optical coherence tomography analysis. Proliferated tunica media were noted in the nicotine injection with balloon overdilation groups and lack of collagen was observed in the tunica media at eight weeks. Quantitative analysis showed increased smooth muscle actin alpha (SMA), cluster of differentiation 68 (CD68), and Krüppel-like factor 4 (KLF4) in the nicotine injection with balloon overdilation groups. Immunohistochemistry results showed CD68-positive cells displayed SMA- and KLF4-positive reactivity in the border zone of the intimal hyperplasia. Our results show that nicotine injection with balloon overdilation can induce atherosclerotic lesions within one month, which can serve as an alternative pig animal model for the development of coronary stents.
Collapse
Affiliation(s)
- Munki Kim
- The Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, 61469, Republic of Korea
| | - Han Byul Kim
- The Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, 61469, Republic of Korea
| | - Dae Sung Park
- The Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, 61469, Republic of Korea.,Korea Cardiovascular Stent Research Institute, Jangsung, 57248, Republic of Korea
| | - Kyung Hoon Cho
- Division of Cardiology of Chonnam, National University Hospital, Cardiovascular Convergence Research Center Nominated By Korea Ministry of Health and Welfare, Gwangju, Republic of Korea
| | - Dae Young Hyun
- Division of Cardiology of Chonnam, National University Hospital, Cardiovascular Convergence Research Center Nominated By Korea Ministry of Health and Welfare, Gwangju, Republic of Korea
| | - Hae Jin Kee
- The Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, 61469, Republic of Korea
| | - Young Joon Hong
- Division of Cardiology of Chonnam, National University Hospital, Cardiovascular Convergence Research Center Nominated By Korea Ministry of Health and Welfare, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- The Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, 61469, Republic of Korea. .,Korea Cardiovascular Stent Research Institute, Jangsung, 57248, Republic of Korea. .,Division of Cardiology of Chonnam, National University Hospital, Cardiovascular Convergence Research Center Nominated By Korea Ministry of Health and Welfare, Gwangju, Republic of Korea.
| |
Collapse
|
18
|
Song L, Zhang D, Guo C, Gu Z, Wang L, Yao YS, Wang H, Zeng Z, Wang W, Yang Y, Bei W, Rong X, Guo J. The traditional Chinese medicine formula Fufang-Zhenzhu-Tiaozhi protects myocardia from injury in diabetic minipigs with coronary heart disease. Biomed Pharmacother 2021; 137:111343. [PMID: 33761594 DOI: 10.1016/j.biopha.2021.111343] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetes mellitus (DM) is a major risk factor for coronary heart disease (CHD). Previous research has reported that the Fufang-Zhenzhu-Tiaozhi (FTZ) formula has obvious effects on the treatment of dyslipidemia and hyperglycemia. In the present study, we intended to establish a convenient DM-CHD model in minipigs and investigated the protective effect of FTZ against myocardial injury and its mechanism. METHODS The DM-CHD model was established by a high-fat/high-sucrose/high-cholesterol diet (HFSCD) combined with balloon injury in the coronary artery. Subsequently, sixteen Wuzhishan minipigs were assigned to three groups: control group, model group, and FTZ group. The model group and FTZ group were given a HFSCD, while the control group was given a normal diet (ND). FTZ was given with meals in the FTZ group. During this time, biochemical parameters, such as total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein (HDL-C), and fasting blood glucose (FBG), were measured by using testing kits. Insulin (INS) was determined by ELISA, and the homeostasis model assessment index of insulin resistance (HOMA-IR) was calculated to evaluate insulin resistance levels. After FTZ administration, the plasma levels of lactate dehydrogenase (LDH), creatine kinase isoenzyme MB (CK-MB), and cardiac troponin I (cTnI) were measured by using ELISA kits to evaluate myocardial injury. Coronary artery stenosis was analyzed by angiographic and HE staining. Myocardial ischemia was assayed with electrocardiogram (ECG). Moreover, cytokines, including interleukin-6 (IL-6), hypersensitive C-reactive protein (hs-CRP), and tumor necrosis factor-alpha (TNF-α), were measured by ELISA kits to assess inflammation. The myocardial tissue was collected, and the pathological morphology was observed by transmission electron microscopy (TEM), HE staining, and Masson staining. Western blots were used to detect the expression of PI3K, AKT, p-AKT, p-NF-κB, and NF-κB. RESULTS A DM-CHD model in minipigs with glucose-lipid metabolism disorder, coronary artery incrassation and myocardial damage was successfully established through balloon injury in the coronary artery combined with HFSCD. FTZ effectively inhibited coronary artery incrassation and protected the myocardium against injury in DM-CHD minipigs. FTZ decreased proinflammatory cytokine levels and upregulated the protein expression of the PI3K/Akt pathway in the myocardium. CONCLUSIONS A novel DM-CHD model in minipigs was successfully established through balloon injury in the coronary artery combined with HFSCD. FTZ has a protective effect against myocardial injury in DM-CHD by inhibiting inflammation and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Lixia Song
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Dongxing Zhang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Caijuan Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Zhanhui Gu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Lexun Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Yu Si Yao
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Hong Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Zhihuan Zeng
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Weixuan Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Yiqi Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Weijian Bei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Xianglu Rong
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory against Metabolic Diseases, China.
| |
Collapse
|
19
|
A novel method for isolation and culture of primary swine gastric epithelial cells. BMC Mol Cell Biol 2021; 22:1. [PMID: 33407092 PMCID: PMC7789315 DOI: 10.1186/s12860-020-00341-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/22/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Culturing primary epithelial cells has a major advantage over tumor-derived or immortalized cell lines as long as their functional phenotype and genetic makeup are mainly maintained. The swine model has shown to be helpful and reliable when used as a surrogate model for human diseases. Several porcine cell lines have been established based on a variety of tissues, which have shown to extensively contribute to the current understanding of several pathologies, especially cancer. However, protocols for the isolation and culture of swine gastric epithelial cells that preserve cell phenotype are rather limited. We aimed to develop a new method for establishing a primary epithelial cell culture from the fundic gland region of the pig stomach. RESULTS Mechanical and enzymatic dissociation of gastric tissue was possible by combining collagenase type I and dispase II, protease inhibitors and antioxidants, which allowed the isolation of epithelial cells from the porcine fundic glands showing cell viability > 90% during the incubation period. Gastric epithelial cells cultured in RPMI 1640, DMEM-HG and DMEM/F12 media did not contribute enough to cell adhesion, cluster formation and cell proliferation. By contrast, William's E medium supplemented with growth factors supports confluency and proliferation of a pure epithelial cell monolayer after 10 days of incubation at 37 °C, 5% CO2. Mucin-producing cell phenotype of primary isolates was confirmed by PAS staining, MUC1 by immunohistochemistry, as well as the expression of MUC1 and MUC20 genes by RT-PCR and cDNA sequencing. Swine gastric epithelial cells also showed origin-specific markers such as cytokeratin cocktail (AE1/AE3) and cytokeratin 18 (CK-18) using immunohistochemical and immunofluorescence methods, respectively. CONCLUSIONS A new method was successfully established for the isolation of primary gastric epithelial cells from the fundic gland zone through a swine model based on a combination of tissue-specific proteases, protease inhibitors and antioxidants after mechanical cell dissociation. The formulation of William's E medium with growth factors for epithelial cells contributes to cell adhesion and preserves functional primary cells phenotype, which is confirmed by mucin production and expression of typical epithelial markers over time.
Collapse
|
20
|
Jové M, Tibau J, Serrano JCE, Berdún R, Rodríguez-Palmero M, Font-i-Furnols M, Cassanyé A, Rodriguez-Mortera R, Sol J, Rassendren H, Fàbrega E, Crescenti A, Castell A, Sabater M, Ortega FJ, Martin-Gari M, Quintanilla R, Puigjaner J, Moreno JA, Prat J, Arola L, Fernández-Real JM, Pamplona R, Portero-Otin M. Molecular phenomics of a high-calorie diet-induced porcine model of prepubertal obesity. J Nutr Biochem 2020; 83:108393. [DOI: 10.1016/j.jnutbio.2020.108393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 03/05/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
|
21
|
Wang D, Xu X, Zhao M, Wang X. Accelerated miniature swine models of advanced atherosclerosis: A review based on morphology. Cardiovasc Pathol 2020; 49:107241. [PMID: 32554057 DOI: 10.1016/j.carpath.2020.107241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/31/2022] Open
Abstract
In order to accelerate development of atherosclerosis(AS) in miniature swine models, varieties of strategies and methods have been explored. In addition to traditional methods such as high cholesterol feeding and balloon injury, new methods such as familial hypercholesterolemia induced by gene editing and intramural injection have been applied in recent years. Although it has been claimed that these methods have successfully aggravated lesion areas and stenosis, lesion features induced by different strategies have shown heterogeneity in morphology. In addition, time consumption, high cost, and unavailability are problems that restrict application of these AS models. Here, we summarize strategies and methods to accelerate AS models and further analyze their values, advantages, and shortcomings.
Collapse
Affiliation(s)
- Dayang Wang
- Cardiovascular Department, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.
| | - Xiaoqing Xu
- Third Department of Neurology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Xian Wang
- Cardiovascular Insititute, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
22
|
Andreadou I, Schulz R, Badimon L, Adameová A, Kleinbongard P, Lecour S, Nikolaou PE, Falcão-Pires I, Vilahur G, Woudberg N, Heusch G, Ferdinandy P. Hyperlipidaemia and cardioprotection: Animal models for translational studies. Br J Pharmacol 2020; 177:5287-5311. [PMID: 31769007 DOI: 10.1111/bph.14931] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Hyperlipidaemia is a well-established risk factor for cardiovascular diseases and therefore, many animal model have been developed to mimic the human abnormal elevation of blood lipid levels. In parallel, extensive research for the alleviation of ischaemia/reperfusion injury has revealed that hyperlipidaemia is a major co-morbidity that attenuates the cardioprotective effect of conditioning strategies (preconditioning, postconditioning and remote conditioning) and that of pharmacological interventions by interfering with cardioprotective signalling pathways. In the present review article, we summarize the existing data on animal models of hypercholesterolaemia (total, low density and HDL abnormalities) and hypertriglyceridaemia used in ischaemia/reperfusion injury and protection from it. We also provide recommendations on preclinical animal models to be used for translations of the cardioprotective strategies into clinical practice. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic.,Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovak Republic
| | - Petra Kleinbongard
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Ines Falcão-Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Nicholas Woudberg
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gerd Heusch
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
23
|
Porcine models for studying complications and organ crosstalk in diabetes mellitus. Cell Tissue Res 2020; 380:341-378. [PMID: 31932949 DOI: 10.1007/s00441-019-03158-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023]
Abstract
The worldwide prevalence of diabetes mellitus and obesity is rapidly increasing not only in adults but also in children and adolescents. Diabetes is associated with macrovascular complications increasing the risk for cardiovascular disease and stroke, as well as microvascular complications leading to diabetic nephropathy, retinopathy and neuropathy. Animal models are essential for studying disease mechanisms and for developing and testing diagnostic procedures and therapeutic strategies. Rodent models are most widely used but have limitations in translational research. Porcine models have the potential to bridge the gap between basic studies and clinical trials in human patients. This article provides an overview of concepts for the development of porcine models for diabetes and obesity research, with a focus on genetically engineered models. Diabetes-associated ocular, cardiovascular and renal alterations observed in diabetic pig models are summarized and their similarities with complications in diabetic patients are discussed. Systematic multi-organ biobanking of porcine models of diabetes and obesity and molecular profiling of representative tissue samples on different levels, e.g., on the transcriptome, proteome, or metabolome level, is proposed as a strategy for discovering tissue-specific pathomechanisms and their molecular key drivers using systems biology tools. This is exemplified by a recent study providing multi-omics insights into functional changes of the liver in a transgenic pig model for insulin-deficient diabetes mellitus. Collectively, these approaches will provide a better understanding of organ crosstalk in diabetes mellitus and eventually reveal new molecular targets for the prevention, early diagnosis and treatment of diabetes mellitus and its associated complications.
Collapse
|
24
|
Hoogendoorn A, den Hoedt S, Hartman EMJ, Krabbendam-Peters I, Te Lintel Hekkert M, van der Zee L, van Gaalen K, Witberg KT, Dorst K, Ligthart JMR, Drouet L, Van der Heiden K, van Lennep JR, van der Steen AFW, Duncker DJ, Mulder MT, Wentzel JJ. Variation in Coronary Atherosclerosis Severity Related to a Distinct LDL (Low-Density Lipoprotein) Profile: Findings From a Familial Hypercholesterolemia Pig Model. Arterioscler Thromb Vasc Biol 2019; 39:2338-2352. [PMID: 31554418 PMCID: PMC6818985 DOI: 10.1161/atvbaha.119.313246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE In an adult porcine model of familial hypercholesterolemia (FH), coronary plaque development was characterized. To elucidate the underlying mechanisms of the observed inter-individual variation in disease severity, detailed lipoprotein profiles were determined. Approach and Results: FH pigs (3 years old, homozygous LDLR R84C mutation) received an atherogenic diet for 12 months. Coronary atherosclerosis development was monitored using serial invasive imaging and histology. A pronounced difference was observed between mildly diseased pigs which exclusively developed early lesions (maximal plaque burden, 25% [23%-34%]; n=5) and advanced-diseased pigs (n=5) which developed human-like, lumen intruding plaques (maximal plaque burden, 69% [57%-77%]) with large necrotic cores, intraplaque hemorrhage, and calcifications. Advanced-diseased pigs and mildly diseased pigs displayed no differences in conventional risk factors. Additional plasma lipoprotein profiling by size-exclusion chromatography revealed 2 different LDL (low-density lipoprotein) subtypes: regular and larger LDL. Cholesterol, sphingosine-1-phosphate, ceramide, and sphingomyelin levels were determined in these LDL-subfractions using standard laboratory techniques and high-pressure liquid chromatography mass-spectrometry analyses, respectively. At 3 months of diet, regular LDL of advanced-diseased pigs contained relatively more cholesterol (LDL-C; regular/larger LDL-C ratio 1.7 [1.3-1.9] versus 0.8 [0.6-0.9]; P=0.008) than mildly diseased pigs, while larger LDL contained more sphingosine-1-phosphate, ceramides, and sphingomyelins. Larger and regular LDL was also found in plasma of 3 patients with homozygous FH with varying LDL-C ratios. CONCLUSIONS In our adult FH pig model, inter-individual differences in atherosclerotic disease severity were directly related to the distribution of cholesterol and sphingolipids over a distinct LDL profile with regular and larger LDL shortly after the diet start. A similar LDL profile was detected in patients with homozygous FH.
Collapse
Affiliation(s)
- Ayla Hoogendoorn
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| | - Sandra den Hoedt
- Department of Internal Medicine, Laboratory of Vascular Medicine, Division of Pharmacology, Vascular & Metabolic Disease (S.d.H., L.v.d.Z., K.D., J.R.v.L., M.T.M.), Erasmus MC, Rotterdam, the Netherlands
| | - Eline M J Hartman
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| | - Ilona Krabbendam-Peters
- Department of Cardiology, Experimental Cardiology (I.K.-P., M.t.L.H., D.J.D.), Erasmus MC, Rotterdam, the Netherlands
| | - Maaike Te Lintel Hekkert
- Department of Cardiology, Experimental Cardiology (I.K.-P., M.t.L.H., D.J.D.), Erasmus MC, Rotterdam, the Netherlands
| | - Leonie van der Zee
- Department of Internal Medicine, Laboratory of Vascular Medicine, Division of Pharmacology, Vascular & Metabolic Disease (S.d.H., L.v.d.Z., K.D., J.R.v.L., M.T.M.), Erasmus MC, Rotterdam, the Netherlands
| | - Kim van Gaalen
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| | - Karen Th Witberg
- Department of Cardiology, Interventional Cardiology (K.T.W., J.M.R.L.), Erasmus MC, Rotterdam, the Netherlands
| | - Kristien Dorst
- Department of Internal Medicine, Laboratory of Vascular Medicine, Division of Pharmacology, Vascular & Metabolic Disease (S.d.H., L.v.d.Z., K.D., J.R.v.L., M.T.M.), Erasmus MC, Rotterdam, the Netherlands
| | - Jurgen M R Ligthart
- Department of Cardiology, Interventional Cardiology (K.T.W., J.M.R.L.), Erasmus MC, Rotterdam, the Netherlands
| | - Ludovic Drouet
- Department of Angiohematology, Hospital Lariboisiere, Paris, France (L.D.)
| | - Kim Van der Heiden
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Laboratory of Vascular Medicine, Division of Pharmacology, Vascular & Metabolic Disease (S.d.H., L.v.d.Z., K.D., J.R.v.L., M.T.M.), Erasmus MC, Rotterdam, the Netherlands
| | - Antonius F W van der Steen
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| | - Dirk J Duncker
- Department of Cardiology, Experimental Cardiology (I.K.-P., M.t.L.H., D.J.D.), Erasmus MC, Rotterdam, the Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Laboratory of Vascular Medicine, Division of Pharmacology, Vascular & Metabolic Disease (S.d.H., L.v.d.Z., K.D., J.R.v.L., M.T.M.), Erasmus MC, Rotterdam, the Netherlands
| | - Jolanda J Wentzel
- From the Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, the Netherlands (A.H., E.M.J.H., K.v.G., K.V.d.H., A.F.W.v.d.S., J.J.W.)
| |
Collapse
|
25
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
26
|
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
|
27
|
Hall S, Agrawal DK. Delivery of viral vectors for gene therapy in intimal hyperplasia and restenosis in atherosclerotic swine. Drug Deliv Transl Res 2018; 8:918-927. [PMID: 28707263 DOI: 10.1007/s13346-017-0409-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases including atherosclerosis are a major financial and health burden globally. Inflammation associated with atherosclerosis results in the development of plaques that can rupture causing thrombosis, stroke, or death. The most widely used treatment for the removal of atherosclerotic plaques is percutaneous transluminal coronary angioplasty (PTCA) with or without stenting. Although this is a safer and minimally invasive method, restenosis and intimal hyperplasia after interventional procedure remains a major hurdle and more refined approaches are needed. Studies in large animal models such as pigs have facilitated a greater understanding of the underlying mechanisms of the disease and provided novel targets for therapeutic intervention. In pre-clinical studies, viral vector gene therapy has emerged as a promising option for the reduction and/or prevention of restenosis and intimal hyperplasia. Although studies in animal models have generated promising results, clinical trials have yet to prove the clinical efficacy of gene therapy in coronary artery diseases. In this review, we examined and critically reviewed the most recent advances in viral vector gene therapy obtained from studies using porcine model of atherosclerosis.
Collapse
Affiliation(s)
- Sannette Hall
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA. .,Department of Clinical and Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, School of Medicine, Creighton University, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
28
|
Fuchs T, Loureiro MDP, Macedo LE, Nocca D, Nedelcu M, Costa-Casagrande TA. Modelos animais na síndrome metabólica. Rev Col Bras Cir 2018; 45:e1975. [PMID: 30379216 DOI: 10.1590/0100-6991e-20181975] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
RESUMO O conhecimento sobre modelos animais para estudo metabólico representa a base da pesquisa nessa área. Este trabalho tem por objetivo revisar os principais modelos animais a serem utilizados no estudo da obesidade e da síndrome metabólica. Para isso, pesquisa no banco de dados Pubmed foi realizada usando as palavras-chave “animal models”, “obesity”, "metabolic syndrome”, e “bariatric surgery”. Várias espécies de animais podem ser usadas para o estudo de distúrbios metabólicos, no entanto, os roedores, tanto modelos monogênicos quanto modelos de obesidade induzida por dieta (DIO), são os animais mais utilizados nessa área. Animais monogênicos são a melhor escolha se apenas um aspecto estiver sendo avaliado. Animais DIO tendem a demonstrar melhor a interação entre doença, ambiente e gene. No entanto, eles ainda não são totalmente eficazes para a compreensão de todos os mecanismos dessa doença.
Collapse
|
29
|
Cai X, She M, Xu M, Chen H, Li J, Chen X, Zheng D, Liu J, Chen S, Zhu J, Xu X, Li R, Li J, Chen S, Yang X, Li H. GLP-1 treatment protects endothelial cells from oxidative stress-induced autophagy and endothelial dysfunction. Int J Biol Sci 2018; 14:1696-1708. [PMID: 30416384 PMCID: PMC6216037 DOI: 10.7150/ijbs.27774] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 08/22/2018] [Indexed: 12/25/2022] Open
Abstract
Endothelial dysfunction and excessively stimulated autophagy, often caused by oxidant injury or inflammation, will lead to atherosclerosis development and progression in diabetes. The aim of this study is to investigate the protective effect of glucagon-like peptide-1 (GLP-1) treatment on preventing oxidative stress-induced endothelial dysfunction and excessively stimulated autophagy. Treatment of endothelial cells with GLP-1 significantly attenuated oxidative stress-induced endothelial dysfunction and autophagy, which was associated with the reduction of intracellular reactive oxygen species (ROS) levels. These protective effects of GLP-1 were likely mediated by reducing phosphorylation of ERK1/2. We further demonstrated that GLP-1 treatment could reverse downregulation of epigenetic factor histone deacetylase 6 (HDAC6), a downstream molecular of the EKR1/2, induced by oxidant injury. In conclusion, our results suggest that GLP-1 produces a protective effect on endothelial cells from oxidant injury by preventing endothelial dysfunction and autophagy, which may be dependent on restoring HDAC6 through a GLP-1R-ERK1/2-dependent manner.
Collapse
Affiliation(s)
- Xiangsheng Cai
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Miaoqin She
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences. Guangzhou, 510660, People's Republic of China
| | - Mingyu Xu
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Huiying Chen
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jingjing Li
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xinglu Chen
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Dianpeng Zheng
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jun Liu
- Dermatology Hospital of Southern Medical University, Guangzhou, 510091, People's Republic of China
| | - Shangliang Chen
- ShenZhen Hospital, Southern Medical University, ShenZhen 518101, People's Republic of China
| | - Jianbin Zhu
- Technology Center, Guangdong Vitalife Bio-tech Co.,LTD, FoShan, 528200, People's Republic of China
| | - Xiaosong Xu
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Ruiying Li
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Jinlong Li
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shaolian Chen
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Xiaorong Yang
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Hongwei Li
- Institute of Biotherapy, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
30
|
Sedding DG, Boyle EC, Demandt JAF, Sluimer JC, Dutzmann J, Haverich A, Bauersachs J. Vasa Vasorum Angiogenesis: Key Player in the Initiation and Progression of Atherosclerosis and Potential Target for the Treatment of Cardiovascular Disease. Front Immunol 2018; 9:706. [PMID: 29719532 PMCID: PMC5913371 DOI: 10.3389/fimmu.2018.00706] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/22/2018] [Indexed: 01/08/2023] Open
Abstract
Plaque microvascularization and increased endothelial permeability are key players in the development of atherosclerosis, from the initial stages of plaque formation to the occurrence of acute cardiovascular events. First, endothelial dysfunction and increased permeability facilitate the entry of diverse inflammation-triggering molecules and particles such as low-density lipoproteins into the artery wall from the arterial lumen and vasa vasorum (VV). Recognition of entering particles by resident phagocytes in the vessel wall triggers a maladaptive inflammatory response that initiates the process of local plaque formation. The recruitment and accumulation of inflammatory cells and the subsequent release of several cytokines, especially from resident macrophages, stimulate the expansion of existing VV and the formation of new highly permeable microvessels. This, in turn, exacerbates the deposition of pro-inflammatory particles and results in the recruitment of even more inflammatory cells. The progressive accumulation of leukocytes in the intima, which trigger proliferation of smooth muscle cells in the media, results in vessel wall thickening and hypoxia, which further stimulates neoangiogenesis of VV. Ultimately, this highly inflammatory environment damages the fragile plaque microvasculature leading to intraplaque hemorrhage, plaque instability, and eventually, acute cardiovascular events. This review will focus on the pivotal roles of endothelial permeability, neoangiogenesis, and plaque microvascularization by VV during plaque initiation, progression, and rupture. Special emphasis will be given to the underlying molecular mechanisms and potential therapeutic strategies to selectively target these processes.
Collapse
Affiliation(s)
- Daniel G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Erin C Boyle
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Jasper A F Demandt
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands.,BHF Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - Jochen Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
31
|
Yuan F, Guo L, Park KH, Woollard JR, Taek-Geun K, Jiang K, Melkamu T, Zang B, Smith SL, Fahrenkrug SC, Kolodgie FD, Lerman A, Virmani R, Lerman LO, Carlson DF. Ossabaw Pigs With a PCSK9 Gain-of-Function Mutation Develop Accelerated Coronary Atherosclerotic Lesions: A Novel Model for Preclinical Studies. J Am Heart Assoc 2018; 7:e006207. [PMID: 29572319 PMCID: PMC5907533 DOI: 10.1161/jaha.117.006207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 01/30/2018] [Indexed: 12/03/2022]
Abstract
BACKGROUND Ossabaw pigs are unique miniature swine with genetic predisposition to develop metabolic syndrome and coronary atherosclerosis after extended periods receiving atherogenic diets. We have hypothesized that transgenic Ossabaw swine expressing chimp PCSK9 (proprotein convertase subtilisin-like/kexin type 9) containing the D374Y gain of function would develop familial hypercholesterolemia and coronary artery plaques more rapidly than Landrace swine with the same transgene. METHODS AND RESULTS Ossabaw and Landrace PCSK9 gain-of-function founders were generated by Sleeping Beauty transposition and cloning. Histopathologic findings in the Ossabaw founder animal showed more advanced plaques and higher stenosis than in the Landrace founder, underscoring the Ossabaw genetic predisposition to atherosclerosis. We chose to further characterize the Ossabaw PCSK9 gain-of-function animals receiving standard or atherogenic diets in a 6-month longitudinal study using computed tomography, magnetic resonance (MR) imaging, intravascular ultrasound, and optical coherence tomography, followed by pathological analysis of atherosclerosis focused on the coronary arteries. The Ossabaw model was consistently hypercholesterolemic, with or without dietary challenge, and by 6 months had consistent and diffuse fibrofatty or fibroatheromatous plaques with necrosis, overlying fibrous caps, and calcification in up to 10% of coronary plaques. CONCLUSIONS The Ossabaw PCSK9 gain-of-function model provides consistent and robust disease development in a time frame that is practical for use in preclinical therapeutic evaluation to drive innovation. Although no animal model perfectly mimics the human condition, this genetic large-animal model is a novel tool for testing therapeutic interventions in the context of developing and advanced coronary artery disease.
Collapse
Affiliation(s)
- Fang Yuan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Liang Guo
- CVPath Institute Inc, Gaithersburg, MD
| | - Kyoung-Ha Park
- Division of Cardiovascular Disease, Hallym University Medical Center, Anyang, Korea
| | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Kwon Taek-Geun
- Heart Center, Konyang University Hospital, Daejeon, South Korea
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | | - Bin Zang
- Program of Scientific Computation, University of Minnesota, Minneapolis, MN
| | | | | | | | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
32
|
Dincer Z, Piccicuto V, Walker UJ, Mahl A, McKeag S. Spontaneous and Drug-induced Arteritis/Polyarteritis in the Göttingen Minipig—Review. Toxicol Pathol 2018; 46:121-130. [DOI: 10.1177/0192623318754791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arteritis/polyarteritis occurs spontaneously in many species used in preclinical toxicology studies. In Göttingen minipigs, arteritis/polyarteritis is an occasionally observed background change. In the minipig, this finding differs in frequency and nature from age-related polyarteritis nodosa in rats or monkeys, and Beagle pain syndrome in dogs. In minipigs, it can be present in a single small- or medium-sized artery of an organ or a few organs and is most commonly recorded in the cardiac and extracardiac blood vessels, vagina, oviduct, rectum, epididymis, spinal cord, pancreas, urinary bladder, kidneys, and stomach. The etiology is unknown although it has been considered in minipigs as well as in rats, dogs, and monkeys to be possibly immune mediated. This background change is important with respect to its nature and distribution in the minipig in order to distinguish it from drug-induced vascular changes, which might occur in similar locations and have similar morphologic features. This review summarizes the morphology, incidence, and predilection sites of arteritis as a spontaneously occurring background change and as a drug-induced vasculopathy in the minipig, and also describes the main aspects to consider when evaluating vascular changes in Göttingen minipig toxicity studies and their human relevance.
Collapse
Affiliation(s)
- Zuhal Dincer
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | - Virginie Piccicuto
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | - Ursula Junker Walker
- Preclinical Safety, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Andreas Mahl
- Preclinical Safety, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Sean McKeag
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| |
Collapse
|
33
|
Peripheral vascular atherosclerosis in a novel PCSK9 gain-of-function mutant Ossabaw miniature pig model. Transl Res 2018; 192:30-45. [PMID: 29175268 PMCID: PMC5811343 DOI: 10.1016/j.trsl.2017.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 10/24/2022]
Abstract
Hypercholesterolemia is a major risk factor for atherosclerosis. Remaining challenges in the management of atherosclerosis necessitate development of animal models that mimic human pathophysiology. We characterized a novel mutant pig model with DNA transposition of D374Y gain-of-function (GOF) cDNA of chimp proprotein convertase subtilisin/kexin type-9 (PCSK9), and tested the hypothesis that it would develop peripheral vascular remodeling and target organ injury in the kidney. Wild-type or PCSK9-GOF Ossabaw miniature pigs fed a standard or atherogenic diet (AD) (n = 7 each) were studied in vivo after 3 and 6 months of diet. Single-kidney hemodynamics and function were studied using multidetector computed tomography and kidney oxygenation by blood oxygen level-dependent magnetic resonance imaging. The renal artery was evaluated by intravascular ultrasound, aortic stiffness by multidetector computed tomography, and kidney stiffness by magnetic resonance elastography. Subsequent ex vivo studies included the renal artery endothelial function and morphology of abdominal aorta, renal, and femoral arteries by histology. Compared with wild type, PCSK9-GOF pigs had elevated cholesterol, triglyceride, and blood pressure levels at 3 and 6 months. Kidney stiffness increased in GOF groups, but aortic stiffness only in GOF-AD. Hypoxia, intrarenal fat deposition, oxidative stress, and fibrosis were observed in both GOF groups, whereas kidney function remained unchanged. Peripheral arteries in GOF groups showed medial thickening and development of atheromatous plaques. Renal endothelial function was impaired only in GOF-AD. Therefore, the PCSK9-GOF mutation induces rapid development of atherosclerosis in peripheral vessels of Ossabaw pigs, which is exacerbated by a high-cholesterol diet. This model may be useful for preclinical studies of atherosclerosis.
Collapse
|
34
|
Bekiares NA, Chen AS, Shanmuganayagam D, Meyers AD, Crenshaw TD, Krueger CG, Reed JD. Effect of Caloric Restriction on Metabolic Dysfunction of Young Rapacz Familial Hypercholesterolemic Swine ( Sus scrofa). Comp Med 2017; 67:508-517. [PMID: 29212583 PMCID: PMC5713166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/08/2017] [Accepted: 04/27/2017] [Indexed: 06/07/2023]
Abstract
The Rapacz familial hypercholesterolemic (FH) swine model is well-characterized and used for studies of both spontaneous and inducible atherosclerosis but has not been used for studies of metabolic dysfunction to date. We examined whether parameters of metabolic syndrome including weight and adiposity, serum cholesterol, and glucoregulatory function could be modulated by restriction of caloric intake in the FH swine. Three groups of FH swine (n = 6 per group) were fed without restriction (AL), 80% of AL caloric intake, or 60% of AL caloric intake for 8.8 ± 0.5 mo beginning 2 wk after weaning. Caloric intake influenced the rate and magnitude of body weight gain and change in adiposity, as determined by dual-emission X-ray absorptiometry. At the conclusion of the study, pigs in the AL group reached a total least-square mean body weight of 94.2 kg and fat mass of 31.1%, whereas those fed 80% AL were 71.6 kg and 24.3% fat, and swine fed 60% AL were 46.1 kg and 14.1% fat. Serum cholesterol was greater in AL than 60% AL pigs at the end of the study. At 10 mo of age, intravenous glucose tolerance testing, performed to assess glucoregulatory function, indicated significant differences in serum glucose clearance profiles and insulin sensitivity between the AL- and 60% AL-fed swine. The AL-fed animals showed almost 5-fold lower insulin sensitivity when compared with animals fed 60% AL caloric intake. These results highlight the value of the FH swine model to study metabolic dysfunction due to changes in caloric intake.
Collapse
Affiliation(s)
- Nell A Bekiares
- Department of Animal Sciences, University of Wisconsin-Madison, Wisconsin
| | - Andrea S Chen
- Department of Animal Sciences, University of Wisconsin-Madison, Wisconsin
| | | | | | - Thomas D Crenshaw
- Department of Animal Sciences, University of Wisconsin-Madison, Wisconsin
| | | | - Jess D Reed
- Department of Animal Sciences, University of Wisconsin-Madison, Wisconsin;,
| |
Collapse
|
35
|
Shim J, Poulsen CB, Hagensen MK, Larsen T, Heegaard PM, Christoffersen C, Bolund L, Schmidt M, Liu Y, Li J, Li R, Callesen H, Bentzon JF, Sørensen CB. Apolipoprotein E Deficiency Increases Remnant Lipoproteins and Accelerates Progressive Atherosclerosis, But Not Xanthoma Formation, in Gene-Modified Minipigs. ACTA ACUST UNITED AC 2017; 2:591-600. [PMID: 30062172 PMCID: PMC6058916 DOI: 10.1016/j.jacbts.2017.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 01/21/2023]
Abstract
APOE-deficient Yucatan minipigs were created by recombinant adeno-associated virus mediated gene targeting in porcine fibroblasts followed by somatic cell nuclear transfer. APOE−/− minipigs displayed increased plasma cholesterol and accumulation of APOB48-containing chylomicron remnants on low fat-diet, which was significantly accentuated upon feeding a high-fat, high-cholesterol diet. APOE−/− minipigs showed accelerated progressive atherosclerosis but not xanthoma formation indicating that remnant lipoproteinemia does not induce early lesions but is atherogenic in pre-existing atherosclerosis.
Deficiency of apolipoprotein E (APOE) causes familial dysbetalipoproteinemia in humans resulting in a higher risk of atherosclerotic disease. In mice, APOE deficiency results in a severe atherosclerosis phenotype, but it is unknown to what extent this is unique to mice. In this study, APOE was targeted in Yucatan minipigs. APOE−/− minipigs displayed increased plasma cholesterol and accumulation of apolipoprotein B-48–containing chylomicron remnants on low-fat diet, which was significantly accentuated upon feeding a high-fat, high-cholesterol diet. APOE−/− minipigs displayed accelerated progressive atherosclerosis but not xanthoma formation. This indicates that remnant lipoproteinemia does not induce early lesions but is atherogenic in pre-existing atherosclerosis.
Collapse
Key Words
- APOB, apolipoprotein B
- APOE, apolipoprotein E
- HFHC, high-fat high-cholesterol
- IDL, intermediate-density lipoprotein
- LAD, left anterior descending (coronary artery)
- LDL, low-density lipoprotein
- LDLR, low-density lipoprotein receptor
- LF, low-fat
- Neo, neomycin
- SMC, smooth muscle cell
- VLDL, very-low-density lipoprotein
- apolipoprotein E
- atherosclerosis
- cDNA, complementary DNA
- pig
- rAAV, recombinant adeno-associated virus
- remnant cholesterol dysbetalipoproteinemia
Collapse
Affiliation(s)
- Jeong Shim
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Christian Bo Poulsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Mette K. Hagensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Torben Larsen
- Department of Animal Science, Aarhus University, Aarhus, Denmark
| | - Peter M.H. Heegaard
- National Veterinary Institute, Technical University of Denmark, Lyngby, Denmark
| | | | - Lars Bolund
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mette Schmidt
- Section for Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ying Liu
- Department of Animal Science, Aarhus University, Aarhus, Denmark
| | - Juan Li
- Department of Animal Science, Aarhus University, Aarhus, Denmark
| | - Rong Li
- Department of Animal Science, Aarhus University, Aarhus, Denmark
| | - Henrik Callesen
- Department of Animal Science, Aarhus University, Aarhus, Denmark
| | - Jacob F. Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Address for correspondence: Dr. Jacob F. Bentzon, CNIC, C/Melchor Fernández Almagro, 28029 Madrid, Spain.
| | - Charlotte B. Sørensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Dr. Charlotte B. Sørensen, Aarhus University, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark.
| |
Collapse
|
36
|
Han JS, Sung JH, Lee SK. Inhibition of Cholesterol Synthesis in HepG2 Cells by GINST-Decreasing HMG-CoA Reductase Expression Via AMP-Activated Protein Kinase. J Food Sci 2017; 82:2700-2705. [DOI: 10.1111/1750-3841.13828] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/16/2017] [Accepted: 06/30/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Joon-Seung Han
- BioTech Research Laboratory; Central Research Inst.; Ilhwa Co., Ltd. Gangdong-gu Seoul 05288 Republic of Korea
| | - Jong Hwan Sung
- BioTech Research Laboratory; Central Research Inst.; Ilhwa Co., Ltd. Gangdong-gu Seoul 05288 Republic of Korea
| | - Seung Kwon Lee
- BioTech Research Laboratory; Central Research Inst.; Ilhwa Co., Ltd. Gangdong-gu Seoul 05288 Republic of Korea
| |
Collapse
|
37
|
Zhao Y, Xiang L, Liu Y, Niu M, Yuan J, Chen H. Atherosclerosis Induced by a High-Cholesterol and High-Fat Diet in the Inbred Strain of the Wuzhishan Miniature Pig. Anim Biotechnol 2017. [PMID: 28636491 DOI: 10.1080/10495398.2017.1322974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Coronary artery disease has a significant genetic predisposition, which mainly results from atherosclerosis. Miniature pig is an excellent model to investigate atherosclerosis. This study investigated whether the occurrence and development of atherosclerosis in the Wuzhishan miniature pigs (WZSPs) that were closely bred 12 generations had better consistency. The WZSPs (n = 9) were fed a high-cholesterol and high-fat diet (HCFD). After continuous feeding, 3 WZSPs each were sacrificed at 6, 8, and 12 months, respectively, and the general clinical manifestations and serological indexes were detected. The pathological changes of the major arteries and main organs were recorded. The results showed WZSPs were quite susceptible to the HCFD. At 6 months, plaque lesions appeared in the abdominal aorta and iliac artery, while at 8 months, they appeared in the coronary artery. At 12 months, atherosclerotic lesions could be found in all major arteries, while lipid core, cholesterol precipitation, and calcium deposition appeared in the most serious sites. The progression of arterial lesions and distribution of the lesions were highly consistent in the pigs. However, apparent variations in serum markers were observed. In conclusion, inbred WZSP is a good model to investigate atherosclerosis and has good predictability for the occurrence and development of the disease.
Collapse
Affiliation(s)
- Yuqiong Zhao
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China
| | - Lei Xiang
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China
| | - Yaqian Liu
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China
| | - Miaomiao Niu
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China
| | - Jifang Yuan
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China
| | - Hua Chen
- a Laboratory Animal Center, Chinese PLA General Hospital , Beijing , PR China.,b State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital , Beijing , PR China
| |
Collapse
|
38
|
Frederiksen SD, Karlskov-Mortensen P, Pant SD, Guerin M, Lesnik P, Jørgensen CB, Cirera S, Bruun CS, Mark T, Fredholm M. Haplotypes on pig chromosome 3 distinguish metabolically healthy from unhealthy obese individuals. PLoS One 2017; 12:e0178828. [PMID: 28570654 PMCID: PMC5453593 DOI: 10.1371/journal.pone.0178828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/19/2017] [Indexed: 01/29/2023] Open
Abstract
We have established a pig resource population specifically designed to elucidate the genetics involved in development of obesity and obesity related co-morbidities by crossing the obesity prone Göttingen Minipig breed with two lean production pig breeds. In this study we have performed genome wide association (GWA) to identify loci with effect on blood lipid levels. The most significantly associated single nucleotide polymorphisms (SNPs) were used for linkage disequilibrium (LD) and haplotype analyses. Three separate haploblocks which influence the ratio between high density lipoprotein cholesterol and total cholesterol (HDL-C/CT), triglycerides (TG) and low density lipoprotein cholesterol (LDL-C) levels respectively were identified on Sus Scrofa chromosome 3 (SSC3). Large additive genetic effects were found for the HDL-C/CT and LDL-C haplotypes. Haplotypes segregating from Göttingen Minipigs were shown to impose a positive effect on blood lipid levels. Thus, the genetic profile of the Göttingen Minipig breed seems to support a phenotype comparable to the metabolic healthy obese (MHO) phenotype in humans.
Collapse
Affiliation(s)
- Simona D. Frederiksen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Karlskov-Mortensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sameer D. Pant
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, Australia
| | - Maryse Guerin
- INSERM UMR_S1166, Integrative Biology of Atherosclerosis Team, Paris, France
| | - Philippe Lesnik
- INSERM UMR_S1166, Integrative Biology of Atherosclerosis Team, Paris, France
| | - Claus B. Jørgensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla S. Bruun
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Mark
- Novo Nordisk, Scandinavia AB, Region Denmark, Maaloev, Denmark
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
39
|
Watson SR, Lessner SM. (Second) Harmonic Disharmony: Nonlinear Microscopy Shines New Light on the Pathology of Atherosclerosis. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2016; 22:589-98. [PMID: 27329310 DOI: 10.1017/s1431927616000842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
There has been increasing interest in second harmonic generation (SHG) imaging approaches for the investigation of atherosclerosis due to the deep penetration and three-dimensional sectioning capabilities of the nonlinear optical microscope. Atherosclerosis involves remodeling or alteration of the collagenous framework in affected vessels. The disease is often characterized by excessive collagen deposition and altered collagen organization. SHG has the capability to accurately characterize collagen structure, which is an essential component in understanding atherosclerotic lesion development and progression. As a structure-based imaging modality, SHG is most impactful in atherosclerosis evaluation in conjunction with other, chemically specific nonlinear optics (NLO) techniques to identify additional components of the lesion. These include the use of coherent anti-Stokes Raman scattering and two-photon excitation fluorescence for studying atherosclerosis burden, and application of stimulated Raman scattering to image cholesterol crystals. However, very few NLO studies have attempted to quantitate differences in control versus atherosclerotic states or to correlate the application to clinical situations. This review highlights the potential of SHG imaging to directly and indirectly describe atherosclerosis as a pathological condition.
Collapse
Affiliation(s)
- Shana R Watson
- Department of Cell Biology and Anatomy,University of South Carolina School of Medicine,Columbia,SC,USA
| | - Susan M Lessner
- Department of Cell Biology and Anatomy,University of South Carolina School of Medicine,Columbia,SC,USA
| |
Collapse
|
40
|
Schomberg DT, Tellez A, Meudt JJ, Brady DA, Dillon KN, Arowolo FK, Wicks J, Rousselle SD, Shanmuganayagam D. Miniature Swine for Preclinical Modeling of Complexities of Human Disease for Translational Scientific Discovery and Accelerated Development of Therapies and Medical Devices. Toxicol Pathol 2016; 44:299-314. [PMID: 26839324 DOI: 10.1177/0192623315618292] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Noncommunicable diseases, including cardiovascular disease, diabetes, chronic respiratory disease, and cancer, are the leading cause of death in the world. The cost, both monetary and time, of developing therapies to prevent, treat, or manage these diseases has become unsustainable. A contributing factor is inefficient and ineffective preclinical research, in which the animal models utilized do not replicate the complex physiology that influences disease. An ideal preclinical animal model is one that responds similarly to intrinsic and extrinsic influences, providing high translatability and concordance of preclinical findings to humans. The overwhelming genetic, anatomical, physiological, and pathophysiological similarities to humans make miniature swine an ideal model for preclinical studies of human disease. Additionally, recent development of precision gene-editing tools for creation of novel genetic swine models allows the modeling of highly complex pathophysiology and comorbidities. As such, the utilization of swine models in early research allows for the evaluation of novel drug and technology efficacy while encouraging redesign and refinement before committing to clinical testing. This review highlights the appropriateness of the miniature swine for modeling complex physiologic systems, presenting it as a highly translational preclinical platform to validate efficacy and safety of therapies and devices.
Collapse
Affiliation(s)
- Dominic T Schomberg
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jennifer J Meudt
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | - Folagbayi K Arowolo
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joan Wicks
- Alizée Pathology, LLC, Thurmont, Maryland, USA
| | | | | |
Collapse
|
41
|
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
|
42
|
Porras AM, Shanmuganayagam D, Meudt JJ, Krueger CG, Hacker TA, Rahko PS, Reed JD, Masters KS. Development of Aortic Valve Disease in Familial Hypercholesterolemic Swine: Implications for Elucidating Disease Etiology. J Am Heart Assoc 2015; 4:e002254. [PMID: 26508741 PMCID: PMC4845146 DOI: 10.1161/jaha.115.002254] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background Familial hypercholesterolemia (FH) is a prevalent hereditary disease associated with increased atherosclerosis and calcific aortic valve disease (CAVD). However, in both FH and non‐FH individuals, the role of hypercholesterolemia in the development of CAVD is poorly understood. This study used Rapacz FH (RFH) swine, an established model of human FH, to investigate the role of hypercholesterolemia alone in the initiation and progression of CAVD. The valves of RFH swine have not previously been examined. Methods and Results Aortic valve leaflets were isolated from wild‐type (0.25‐ and 1‐year‐old) and RFH (0.25‐, 1‐, 2‐, and 3‐year‐old) swine. Adult RFH animals exhibited numerous hallmarks of early CAVD. Significant leaflet thickening was found in adult RFH swine, accompanied by extensive extracellular matrix remodeling, including proteoglycan enrichment, collagen disorganization, and elastin fragmentation. Increased lipid oxidation and infiltration of macrophages were also evident in adult RFH swine. Intracardiac echocardiography revealed mild aortic valve sclerosis in some of the adult RFH animals, but unimpaired valve function. Microarray analysis of valves from adult versus juvenile RFH animals revealed significant upregulation of inflammation‐related genes, as well as several commonalities with atherosclerosis and overlap with human CAVD. Conclusions Adult RFH swine exhibited several hallmarks of early human CAVD, suggesting potential for these animals to help elucidate CAVD etiology in both FH and non‐FH individuals. The development of advanced atherosclerotic lesions, but only early‐stage CAVD, in RFH swine supports the hypothesis of an initial shared disease process, with additional stimulation necessary for further progression of CAVD.
Collapse
Affiliation(s)
- Ana M. Porras
- Department of Biomedical EngineeringUniversity of Wisconsin–MadisonMadisonWI
| | | | - Jennifer J. Meudt
- Department of Animal SciencesUniversity of Wisconsin–MadisonMadisonWI
| | | | - Timothy A. Hacker
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of Wisconsin–MadisonMadisonWI
| | - Peter S. Rahko
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of Wisconsin–MadisonMadisonWI
| | - Jess D. Reed
- Department of Animal SciencesUniversity of Wisconsin–MadisonMadisonWI
| | - Kristyn S. Masters
- Department of Biomedical EngineeringUniversity of Wisconsin–MadisonMadisonWI
| |
Collapse
|
43
|
Shim J, Al-Mashhadi RH, Sørensen CB, Bentzon JF. Large animal models of atherosclerosis - new tools for persistent problems in cardiovascular medicine. J Pathol 2015; 238:257-66. [DOI: 10.1002/path.4646] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 11/06/2022]
Affiliation(s)
- J Shim
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - RH Al-Mashhadi
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - CB Sørensen
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - JF Bentzon
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III; Madrid Spain
| |
Collapse
|
44
|
Ludvigsen TP, Kirk RK, Christoffersen BØ, Pedersen HD, Martinussen T, Kildegaard J, Heegaard PMH, Lykkesfeldt J, Olsen LH. Göttingen minipig model of diet-induced atherosclerosis: influence of mild streptozotocin-induced diabetes on lesion severity and markers of inflammation evaluated in obese, obese and diabetic, and lean control animals. J Transl Med 2015; 13:312. [PMID: 26394837 PMCID: PMC4580291 DOI: 10.1186/s12967-015-0670-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/11/2015] [Indexed: 01/01/2023] Open
Abstract
Background From a pharmacological perspective, readily-available, well-characterized animal models of cardiovascular disease, including relevant in vivo markers of atherosclerosis are important for evaluation of novel drug candidates. Furthermore, considering the impact of diabetes mellitus on atherosclerosis in human patients, inclusion of this disease aspect in the characterization of a such model, is highly relevant. The objective of this study was to evaluate the effect of mild streptozotocin-induced diabetes on ex- and in vivo end-points in a diet-induced atherosclerotic minipig model. Methods Castrated male Göttingen minipigs were fed standard chow (CD), atherogenic diet alone (HFD) or with superimposed mild streptozotocin-induced diabetes (HFD-D). Circulating markers of inflammation (C-reactive protein (CRP), oxidized low-density lipoprotein (oxLDL), plasminogen activator inhibitor-1, lipid and glucose metabolism were evaluated together with coronary and aortic atherosclerosis after 22 or 43 diet-weeks. Group differences were evaluated by analysis of variance for parametric data and Kruskal–Wallis test for non-parametric data. For qualitative assessments, Fisher’s exact test was applied. For all analyses, p < 0.05 was considered statistically significant. Results Overall, HFD and HFD-D displayed increased CRP, oxLDL and lipid parameters compared to CD at both time points. HFD-D displayed impaired glucose metabolism as compared to HFD and CD. Advanced atherosclerotic lesions were observed in both coronary arteries and aorta of HFD and HFD-D, with more advanced plaque findings in the aorta but without differences in lesion severity or distribution between HFD and HFD-D. Statistically, triglyceride was positively (p = 0.0039), and high-density lipoprotein negatively (p = 0.0461) associated with aortic plaque area. Conclusions In this model, advanced coronary and aortic atherosclerosis was observed, with increased levels of inflammatory markers, clinically relevant to atherosclerosis. No effect of mild streptozotocin-induced diabetes was observed on plaque area, lesion severity or inflammatory markers. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0670-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Trine Pagh Ludvigsen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg, Denmark. .,GLP-1 and Obesity Pharmacology - PK/PD, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark.
| | - Rikke Kaae Kirk
- Histology & Imaging, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark.
| | | | - Henrik Duelund Pedersen
- GLP-1 and Obesity Pharmacology - PK/PD, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark.
| | - Torben Martinussen
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, Postbox 1014 KBH K, Copenhagen, Denmark.
| | - Jonas Kildegaard
- Clamp Competency Center, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark.
| | - Peter M H Heegaard
- National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870, Frederiksberg, Denmark.
| | - Jens Lykkesfeldt
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg, Denmark.
| | - Lisbeth Høier Olsen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg, Denmark.
| |
Collapse
|
45
|
Site-Specific Secretome Map Evidences VSMC-Related Markers of Coronary Atherosclerosis Grade and Extent in the Hypercholesterolemic Swine. DISEASE MARKERS 2015; 2015:465242. [PMID: 26379359 PMCID: PMC4561865 DOI: 10.1155/2015/465242] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/29/2015] [Accepted: 06/23/2015] [Indexed: 12/20/2022]
Abstract
A major drawback in coronary atherosclerosis (ATS) research is the difficulty of investigating early phase of plaque growth and related features in the clinical context. In this study, secreted proteins from atherosclerotic coronary arteries in a hypercholesterolemic swine model were characterized by a proteomics approach and their expression was correlated to site-specific ATS stage and extent. A wide coronary artery map of secreted proteins has been obtained in high fat (HF) diet induced ATS swine model and a significantly different expression of many proteins related to vascular smooth muscle cell (VSMC) activation/migration has been identified. Significant associations with ATS stage of HF coronary lesions were found for several VSMC-derived proteins and validated for chitinase 3 like protein 1 (CHI3L1) by tissue immunoexpression. A direct correlation (R(2) = 0.85) was evidenced with intima to media thickness ratio values and ELISA confirmed the higher blood concentrations of CHI3L1 in HF cases. These findings confirmed the pivotal role of VSMCs in coronary plaque development and demonstrated a strong site-specific relation between VSMC-secreted CHI3L1 and lesion grade, suggesting that this protein could be proposed as a useful biomarker for diagnosing and staging of atherosclerotic lesions in coronary artery disease.
Collapse
|
46
|
Abstract
Coronary angioplasty initially employed balloon dilatation only. This technique revolutionized the treatment of coronary artery disease, although outcomes were compromised by acute vessel closure, late constrictive remodeling, and restenosis due to neointimal proliferation. These processes were studied in animal models, which contributed to understanding the biology of endovascular arterial injury. Coronary stents overcome acute recoil, with improvements in the design and metallurgy since then, leading to the development of drug-eluting stents and bioresorbable scaffolds. These devices now undergo computer modeling and benchtop and animal testing before evaluation in clinical trials. Animal models, including rabbit, sheep, dog and pig are available, all with individual benefits and limitations. In smaller mammals, such as mouse and rabbit, the target for stenting is generally the aorta; whereas in larger animals, such as the pig, it is generally the coronary artery. The pig coronary stenting model is a gold-standard for evaluating safety; but insights into biomechanical properties, the biology of stenting, and efficacy in controlling neointimal proliferation can also be gained. Intra-coronary imaging modalities such as intravascular ultrasound and optical coherence tomography allow precise serial evaluation in vivo, and recent developments in genetically modified animal models of atherosclerosis provide realistic test beds for future stents and scaffolds.
Collapse
|
47
|
Mahajan S, Chauhan P, Subramani SK, Anand A, Borole D, Goswamy H, GBKS P. Evaluation of “GSPF kwath”: A Gymnema sylvestre-containing polyherbal formulation for the treatment of human type 2 diabetes mellitus. Eur J Integr Med 2015. [DOI: 10.1016/j.eujim.2015.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
48
|
Reduction of histopathological images through a decrease in H2O2 levels in diabetic rats with polysaccharide peptides. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.bgm.2014.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
49
|
Ludvigsen TP, Wiinberg N, Jensen CJ, Callesen AT, Andersen RW, Jørgensen ASH, Christoffersen BØ, Pedersen HD, Moesgaard SG, Olsen LH. Noninvasive assessment of pulse-wave velocity and flow-mediated vasodilation in anesthetized Göttingen minipigs. Comp Med 2014; 64:471-477. [PMID: 25527028 PMCID: PMC4275083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/02/2014] [Accepted: 05/08/2014] [Indexed: 06/04/2023]
Abstract
Few methods for noninvasive assessment of arterial stiffness and endothelial dysfunction in porcine models are available. The aim of this study was to evaluate methods for assessment of arterial stiffness and endothelial dysfunction in anesthetized Göttingen minipigs. Pulse-wave velocity (PWV) was assessed in male Göttingen minipigs (n = 8; age approximately 60 wk) by using applanation tonometry of the carotid and femoral arteries. In addition, flow-mediated vasodilation (FMD) was assessed by using vascular ultrasonography of the brachial artery to evaluate endothelial dysfunction. To evaluate the reproducibility of the methods, minipigs were anesthetized by intravenous infusion of ketamine and midazolam and examined every other day for a total of 3 trials. Neither examination day nor systolic, diastolic, or mean arterial blood pressure statistically influenced PWV or FMD. The median interexamination coefficient of variation was 17% for PWV and 59% for FMD. Measured values of PWV corresponded largely to those in clinically healthy humans, but FMD values were lower than expected for lean, young animals. Although the ketamine-midazolam anesthesia we used has been associated with minor hemodynamic effects in vivo, in vitro studies suggest that both drugs are vasodilatory. Therefore anesthesia might have influenced the endothelial response, contributing to the modest FMD response and the concurrent high coefficients of variation that we noted. We conclude that PWV—but not FMD—showed acceptable interexamination variation for its potential application in porcine models.
Collapse
Affiliation(s)
- Trine P Ludvigsen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark; Novo Nordisk A/S, Måløv, Denmark
| | - Niels Wiinberg
- Department of Clinical Physiology and Nuclear Medicine, Frederiksberg Hospital, Frederiksberg, Denmark
| | - Christina J Jensen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Annemette T Callesen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Regitze W Andersen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Anne Sofie H Jørgensen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | | | | | | | - Lisbeth H Olsen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
50
|
Hamada N, Fujimichi Y, Iwasaki T, Fujii N, Furuhashi M, Kubo E, Minamino T, Nomura T, Sato H. Emerging issues in radiogenic cataracts and cardiovascular disease. JOURNAL OF RADIATION RESEARCH 2014; 55:831-46. [PMID: 24824673 PMCID: PMC4202294 DOI: 10.1093/jrr/rru036] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 04/01/2014] [Accepted: 04/06/2014] [Indexed: 05/26/2023]
Abstract
In 2011, the International Commission on Radiological Protection issued a statement on tissue reactions (formerly termed non-stochastic or deterministic effects) to recommend lowering the threshold for cataracts and the occupational equivalent dose limit for the crystalline lens of the eye. Furthermore, this statement was the first to list circulatory disease (cardiovascular and cerebrovascular disease) as a health hazard of radiation exposure and to assign its threshold for the heart and brain. These changes have stimulated various discussions and may have impacts on some radiation workers, such as those in the medical sector. This paper considers emerging issues associated with cataracts and cardiovascular disease. For cataracts, topics dealt with herein include (i) the progressive nature, stochastic nature, target cells and trigger events of lens opacification, (ii) roles of lens protein denaturation, oxidative stress, calcium ions, tumor suppressors and DNA repair factors in cataractogenesis, (iii) dose rate effect, radiation weighting factor, and classification systems for cataracts, and (iv) estimation of the lens dose in clinical settings. Topics for cardiovascular disease include experimental animal models, relevant surrogate markers, latency period, target tissues, and roles of inflammation and cellular senescence. Future research needs are also discussed.
Collapse
Affiliation(s)
- Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 2-11-1 Iwado-kita, Komae, Tokyo 201-8511, Japan
| | - Yuki Fujimichi
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 2-11-1 Iwado-kita, Komae, Tokyo 201-8511, Japan
| | - Toshiyasu Iwasaki
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 2-11-1 Iwado-kita, Komae, Tokyo 201-8511, Japan
| | - Noriko Fujii
- Kyoto University Research Reactor Institute (KURRI), 2 Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido 060-8543, Japan
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, 1-1 Daigaku, Kahoku, Ishikawa 920-0293, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, 1-754 Asahimachidori, Chuo-ku, Niigata 951-8510, Japan
| | - Takaharu Nomura
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 2-11-1 Iwado-kita, Komae, Tokyo 201-8511, Japan
| | - Hitoshi Sato
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami, Inashiki, Ibaraki 300-0394, Japan
| |
Collapse
|