1
|
Bamezai S, Zhang Y, Kumari M, Lotfi M, Alsaigh T, Luo L, Kumar GS, Wang F, Ye J, Puri M, Manchanda R, Paluri S, Adkar SS, Kojima Y, Ingelsson A, Bell CF, Lopez NG, Fu C, Choi RB, Miller Z, Barrios L, Walsh S, Ahmad F, Maegdefessel L, Smith BR, Leeper NJ. Pro-efferocytic nanotherapies reduce vascular inflammation without inducing anemia in a large animal model of atherosclerosis. Nat Commun 2024; 15:8034. [PMID: 39271657 PMCID: PMC11399336 DOI: 10.1038/s41467-024-52005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Atherosclerosis is an inflammatory disorder responsible for cardiovascular disease. Reactivation of efferocytosis, the phagocytic removal of cells by macrophages, has emerged as a translational target for atherosclerosis. Systemic blockade of the key 'don't-eat-me' molecule, CD47, triggers the engulfment of apoptotic vascular tissue and potently reduces plaque burden. However, it also induces red blood cell clearance, leading to anemia. To overcome this, we previously developed a macrophage-specific nanotherapy loaded with a chemical inhibitor that promotes efferocytosis. Because it was found to be safe and effective in murine studies, we aimed to advance our nanoparticle into a porcine model of atherosclerosis. Here, we demonstrate that production can be scaled without impairing nanoparticle function. At an early stage of disease, we find our nanotherapy reduces apoptotic cell accumulation and inflammation in the atherosclerotic lesion. Notably, this therapy does not induce anemia, highlighting the translational potential of targeted macrophage checkpoint inhibitors.
Collapse
Affiliation(s)
- Sharika Bamezai
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yapei Zhang
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA
| | - Manisha Kumari
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA
| | - Mozhgan Lotfi
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tom Alsaigh
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lingfeng Luo
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Gayatri Suresh Kumar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Fudi Wang
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Madhu Puri
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA
| | - Romila Manchanda
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA
| | - Sesha Paluri
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA
| | - Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alice Ingelsson
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Caitlin F Bell
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicolas G Lopez
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Changhao Fu
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan B Choi
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zach Miller
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Leo Barrios
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Susan Walsh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Ferhaan Ahmad
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University, Munich, Germany
- German Center for Cardiovascular Research, partner site Munich Heart Alliance, Berlin, Germany
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA.
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
2
|
Kim JH, Shim J, Ko N, Kim HJ, Lee Y, Choi K. Analysis of production efficiency of cloned transgenic Yucatan miniature pigs according to recipient breeds with embryo transfer conditions. Theriogenology 2024; 218:193-199. [PMID: 38330863 DOI: 10.1016/j.theriogenology.2024.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/05/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
The purpose of this study was to compare the efficiency of the production of cloned transgenic Yucatan miniature pigs (YMPs) using two recipient breeds, i.e., YMPs and domestic pigs (DPs), under various embryo transfer conditions. We initially assessed the in vitro developmental competence of embryos obtained via somatic cell nuclear transfer (SCNT) from three different transgenic donor cells. No difference was observed among the three groups regarding developmental competence. Furthermore, the cloning efficiency remained consistent among the three groups after the transfer of the SCNT embryos to each surrogate mother. Subsequently, to compare the efficiency of the production of cloned transgenic YMPs between the two recipient breeds using varying parameters, including ovulation status (preovulation and postovulation), duration of in vitro culture (IVC) (incubated within 24 h and 24-48 h), and the number of transferred SCNT embryos (less than and more than 300), we assessed the pregnancy rates, delivery rates, mean offspring counts, and cloning efficiency. Regarding the ovulation status, YMPs exhibited higher pregnancy rates, delivery rates, and cloning efficiency compared with DPs in both statuses. Moreover, the pregnancy rates, delivery rates, and cloning efficiency were affected by the ovulation status in DPs, but not in YMPs. The comparison of IVC duration between groups revealed that YMPs had higher pregnancy rates vs. DPs in both conditions. SCNT embryos cultured for 24-48 h in YMPs yielded higher delivery rates and cloning efficiency compared with those cultured for less than 24 h in DPs. Finally, the analysis based on the number of transferred SCNT embryos showed that both the pregnancy and delivery rates were higher in YMPs vs. DPs. However, the highest average number of offspring was obtained when more than 300 SCNT embryos were transferred into DPs, whereas the cloning efficiency was higher in YMPs vs. DPs. Our results suggest that YMPs are more suitable recipients than are DPs under various conditions for the production of cloned transgenic YMPs.
Collapse
Affiliation(s)
- Jun-Hyeong Kim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Joohyun Shim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Nayoung Ko
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Hyoung-Joo Kim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Yongjin Lee
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Kimyung Choi
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea.
| |
Collapse
|
3
|
Wang X, Liu TX, Zhang Y, Xu LW, Yuan SL, Cui AL, Guo WW, Wang YF, Yang SM, Zhao JG. Genetically modified pigs: Emerging animal models for hereditary hearing loss. Zool Res 2024; 45:284-291. [PMID: 38485498 PMCID: PMC11017082 DOI: 10.24272/j.issn.2095-8137.2023.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 03/19/2024] Open
Abstract
Hereditary hearing loss (HHL), a genetic disorder that impairs auditory function, significantly affects quality of life and incurs substantial economic losses for society. To investigate the underlying causes of HHL and evaluate therapeutic outcomes, appropriate animal models are necessary. Pigs have been extensively used as valuable large animal models in biomedical research. In this review, we highlight the advantages of pig models in terms of ear anatomy, inner ear morphology, and electrophysiological characteristics, as well as recent advancements in the development of distinct genetically modified porcine models of hearing loss. Additionally, we discuss the prospects, challenges, and recommendations regarding the use pig models in HHL research. Overall, this review provides insights and perspectives for future studies on HHL using porcine models.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Tian-Xia Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Liang-Wei Xu
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuo-Long Yuan
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing 100853, China
| | - A-Long Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230022, China
| | - Wei-Wei Guo
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yan-Fang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shi-Ming Yang
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing 100853, China. E-mail:
| | - Jian-Guo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China. E-mail:
| |
Collapse
|
4
|
Srivastava RAK. New opportunities in the management and treatment of refractory hypercholesterolemia using in vivo CRISPR-mediated genome/base editing. Nutr Metab Cardiovasc Dis 2023; 33:2317-2325. [PMID: 37805309 DOI: 10.1016/j.numecd.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 10/09/2023]
Abstract
AIMS Refractory hypercholesterolemia (RH), caused primarily by the loss-of-function mutation of LDL receptor (LDLR) gene seen in HoFH and HeFH patients, remains a major risk factor for atherosclerotic cardiovascular disease (ASCVD). Statin and ezetimibe combination therapy lower circulating LDL by 30% in HoFH patients. PCSK9 mAB, being an LDLR-dependent therapy, is not effective in HoFH, but lowers LDL by 25% in HeFH patients. A maximum reduction of 50% was noted in HoFH patients treated with ANGPTL3 mAB, which was not enough to achieve therapeutic goal of LDL. Therefore, new approaches are warranted to offer hopes to individuals intolerant to higher dose statins and not able to achieve recommended LDL level. DATA SYNTHESIS New approaches to lower LDL include gene therapy and gene editing. AAV-based gene therapy has shown encouraging results in animal models. Using CRISPR/Cas9-mediated genome/base editing, gain of function and loss of function have been successfully done in animal models. Recent progress in the refinement of genome/base editing has overcome the issues of off-target mutagenesis with ∼1% mutagenesis in case of PCSK9 and almost no off-target mutagenesis in inactivating ANGPTL3 in animal models showing 50% reduction in cholesterol. Current approaches using CRISPR-Cas9 genome/base editing targeting LDLR-dependent and LDLR-independent pathways are underway. CONCLUSIONS The new information on gain of LDLR function and inactivation of ANGPTL3 together with developments in genome/base editing technology to overcome off-target insertion and deletion mutagenesis offer hope to refractory hypercholesterolemic individuals who are at a higher risk of developing ASCVD.
Collapse
|
5
|
Cai L, Hyun SH, Kim E. Stem cell factor's role in enhancing the quality of fertilized and cloned porcine embryos for improved embryonic stem cell derivation. Front Vet Sci 2023; 10:1285530. [PMID: 38033636 PMCID: PMC10687439 DOI: 10.3389/fvets.2023.1285530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Stem cell factor (SCF), a cytokine growth factor, is expressed in various tissues of the male and female reproductive organs, including the testis, ovary, and endometrium. Its primary function involves cell survival, differentiation, and proliferation, achieved through its binding to the c-kit receptor. This study aimed to scrutinize the effects of SCF treatment during in vitro culture (IVC) on both the developmental potential and the efficiency of establishing embryonic stem cells (ESCs) from fertilized and cloned porcine embryos. The rates of cleavage and blastocyst formation exhibited no significant differences between fertilized and cloned embryos, even with the addition of SCF. However, it's worth noting that embryos cloned with Cloud eGFP as donor cells demonstrated notably increased rates of hatched blastocysts when treated with SCF, and this increase was statistically significant (p < 0.05). Furthermore, following the complete dissection of the blastocysts, although there was no significant difference in the SCF-treated group, the area of expansion was significantly reduced (p < 0.01) in the group treated with the antagonistic blocker (ACK2) compared to both the control and SCF-treated groups. These outcomes suggest that the SCF/c-kit signaling pathway might play a pivotal role in embryo implantation. As anticipated, the efficiency of deriving ESCs was significantly higher (p < 0.01) in the group subjected to SCF treatment (12.82 ± 1.02%) compared to the control group (5.41 ± 2.25%). In conclusion, this study highlights the crucial role of SCF in enhancing the quality of porcine embryos, a vital step in obtaining high-quality ESCs.
Collapse
Affiliation(s)
- Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
6
|
Burke-Kleinman J, Gotlieb AI. Progression of Arterial Vasa Vasorum from Regulator of Arterial Homeostasis to Promoter of Atherogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1468-1484. [PMID: 37356574 DOI: 10.1016/j.ajpath.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023]
Abstract
The vasa vasorum (vessels of vessels) are a dynamic microvascular system uniquely distributed to maintain physiological homeostasis of the artery wall by supplying nutrients and oxygen to the outer layers of the artery wall, adventitia, and perivascular adipose tissue, and in large arteries, to the outer portion of the medial layer. Vasa vasorum endothelium and contractile mural cells regulate direct access of bioactive cells and factors present in both the systemic circulation and the arterial perivascular adipose tissue and adventitia to the artery wall. Experimental and human data show that proatherogenic factors and cells gain direct access to the artery wall via the vasa vasorum and may initiate, promote, and destabilize the plaque. Activation and growth of vasa vasorum occur in all blood vessel layers primarily by angiogenesis, producing fragile and permeable new microvessels that may cause plaque hemorrhage and fibrous cap rupture. Ironically, invasive therapies, such as angioplasty and coronary artery bypass grafting, injure the vasa vasorum, leading to treatment failures. The vasa vasorum function both as a master integrator of arterial homeostasis and, once perturbed or injured, as a promotor of atherogenesis. Future studies need to be directed at establishing reliable in vivo and in vitro models to investigate the cellular and molecular regulation of the function and dysfunction of the arterial vasa vasorum.
Collapse
Affiliation(s)
- Jonah Burke-Kleinman
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Sato A, Tsukiyama T, Komeno M, Iwatani C, Tsuchiya H, Kawamoto I, Murase M, Nakagawa T, Itagaki I, Seita Y, Matsumoto S, Nakaya M, Shimizu A, Yamada A, Ema M, Ogita H. Generation of a familial hypercholesterolemia model in non-human primate. Sci Rep 2023; 13:15649. [PMID: 37730951 PMCID: PMC10511719 DOI: 10.1038/s41598-023-42763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited autosomal dominant disorder that is associated with a high plasma level of low-density lipoprotein (LDL) cholesterol, leading to an increased risk of cardiovascular diseases. To develop basic and translational research on FH, we here generated an FH model in a non-human primate (cynomolgus monkeys) by deleting the LDL receptor (LDLR) gene using the genome editing technique. Six LDLR knockout (KO) monkeys were produced, all of which were confirmed to have mutations in the LDLR gene by sequence analysis. The levels of plasma cholesterol and triglyceride were quite high in the monkeys, and were similar to those in FH patients with homozygous mutations in the LDLR gene. In addition, periocular xanthoma was observed only 1 year after birth. Lipoprotein profile analysis showed that the plasma very low-density lipoprotein and LDL were elevated, while the plasma high density lipoprotein was decreased in LDLR KO monkeys. The LDLR KO monkeys were also strongly resistant to medications for hypercholesterolemia. Taken together, we successfully generated a non-human primate model of hypercholesterolemia in which the phenotype is similar to that of homozygous FH patients.
Collapse
Affiliation(s)
- Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Tomoyuki Tsukiyama
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masahiro Komeno
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Kawamoto
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Mitsuru Murase
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Nakagawa
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Iori Itagaki
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Masataka Nakaya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Atsushi Yamada
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Masatsugu Ema
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
8
|
Swier VJ, White KA, Johnson TB, Wang X, Han J, Pearce DA, Singh R, Drack AV, Pfeifer W, Rogers CS, Brudvig JJ, Weimer JM. A novel porcine model of CLN3 Batten disease recapitulates clinical phenotypes. Dis Model Mech 2023; 16:dmm050038. [PMID: 37305926 PMCID: PMC10434985 DOI: 10.1242/dmm.050038] [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: 12/09/2022] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Mouse models of CLN3 Batten disease, a rare lysosomal storage disorder with no cure, have improved our understanding of CLN3 biology and therapeutics through their ease of use and a consistent display of cellular pathology. However, the translatability of murine models is limited by disparities in anatomy, body size, life span and inconsistent subtle behavior deficits that can be difficult to detect in CLN3 mutant mouse models, thereby limiting their use in preclinical studies. Here, we present a longitudinal characterization of a novel miniswine model of CLN3 disease that recapitulates the most common human pathogenic variant, an exon 7-8 deletion (CLN3Δex7/8). Progressive pathology and neuron loss is observed in various regions of the CLN3Δex7/8 miniswine brain and retina. Additionally, mutant miniswine present with retinal degeneration and motor abnormalities, similar to deficits seen in humans diagnosed with the disease. Taken together, the CLN3Δex7/8 miniswine model shows consistent and progressive Batten disease pathology, and behavioral impairment mirroring clinical presentation, demonstrating its value in studying the role of CLN3 and safety/efficacy of novel disease-modifying therapeutics.
Collapse
Affiliation(s)
- Vicki J. Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Katherine A. White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tyler B. Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | | | - Jimin Han
- Department of Ophthalmology, Center for Visual Science, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David A. Pearce
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Ruchira Singh
- Department of Ophthalmology, Center for Visual Science, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Arlene V. Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- University of Iowa Institute for Vision Research, Iowa City, IA 52242, USA
| | - Wanda Pfeifer
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | - Jon J. Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
9
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
10
|
Zhou S, Lenk LJ, Gao Y, Wang Y, Zhao X, Pan M, Huang S, Sun K, Kalds P, Luo Q, Lillico S, Sonstegard T, Scholl UI, Ma B, Petersen B, Chen Y, Wang X. Generation of sheep with defined FecB B and TBXT mutations and porcine blastocysts with KCNJ5 G151R/+ mutation using prime editing. BMC Genomics 2023; 24:313. [PMID: 37308830 DOI: 10.1186/s12864-023-09409-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Rewriting the genomes of living organisms has been a long-standing aim in the biological sciences. The revelation of the CRISPR/Cas9 technology has revolutionized the entire biological field. Since its emergence, this technology has been widely applied to induce gene knockouts, insertions, deletions, and base substitutions. However, the classical version of this system was imperfect for inducing or correcting desired mutations. A subsequent development generated more advanced classes, including cytosine and adenine base editors, which can be used to achieve single nucleotide substitutions. Nevertheless, these advanced systems still suffer from several limitations, such as the inability to edit loci without a suitable PAM sequence and to induce base transversions. On the other hand, the recently emerged prime editors (PEs) can achieve all possible single nucleotide substitutions as well as targeted insertions and deletions, which show promising potential to alter and correct the genomes of various organisms. Of note, the application of PE to edit livestock genomes has not been reported yet. RESULTS In this study, using PE, we successfully generated sheep with two agriculturally significant mutations, including the fecundity-related FecBB p.Q249R and the tail length-related TBXT p.G112W. Additionally, we applied PE to generate porcine blastocysts with a biomedically relevant point mutation (KCNJ5 p.G151R) as a porcine model of human primary aldosteronism. CONCLUSIONS Our study demonstrates the potential of the PE system to edit the genomes of large animals for the induction of economically desired mutations and for modeling human diseases. Although prime-edited sheep and porcine blastocysts could be generated, the editing frequencies are still unsatisfactory, highlighting the need for optimizations in the PE system for efficient generation of large animals with customized traits.
Collapse
Affiliation(s)
- Shiwei Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Laura Johanna Lenk
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Yawei Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yuhui Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaoe Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Menghao Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Shuhong Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Kexin Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Peter Kalds
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
- Department of Animal and Poultry Production, Faculty of Environmental Agricultural Sciences, Arish University, El-Arish, 45511, Egypt
| | - Qi Luo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Simon Lillico
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | | | - Ute I Scholl
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10115, Berlin, Germany
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Bjoern Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany.
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs, Yangling, 712100, China.
| | - Xiaolong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs, Yangling, 712100, China.
| |
Collapse
|
11
|
Comment on: Premature delivery in the domestic sow in response to in utero delivery of AAV9 to fetal piglets. Gene Ther 2023; 30:232-235. [PMID: 36918654 DOI: 10.1038/s41434-023-00395-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
|
12
|
Dong M, Liu J, Liu C, Wang H, Sun W, Liu B. CRISPR/CAS9: A promising approach for the research and treatment of cardiovascular diseases. Pharmacol Res 2022; 185:106480. [PMID: 36191879 DOI: 10.1016/j.phrs.2022.106480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
Abstract
The development of gene-editing technology has been one of the biggest advances in biomedicine over the past two decades. Not only can it be used as a research tool to build a variety of disease models for the exploration of disease pathogenesis at the genetic level, it can also be used for prevention and treatment. This is done by intervening with the expression of target genes and carrying out precise molecular targeted therapy for diseases. The simple and flexible clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene-editing technology overcomes the limitations of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs). For this reason, it has rapidly become a preferred method for gene editing. As a new gene intervention method, CRISPR/Cas9 has been widely used in the clinical treatment of tumours and rare diseases; however, its application in the field of cardiovascular diseases is currently limited. This article reviews the application of the CRISPR/Cas9 editing technology in cardiovascular disease research and treatment, and discusses the limitations and prospects of this technology.
Collapse
Affiliation(s)
- Mengying Dong
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Jiangen Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People's Hospital, 33 Wenyi Road, ShenYang, China, 110016
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| |
Collapse
|
13
|
Hou N, Du X, Wu S. Advances in pig models of human diseases. Animal Model Exp Med 2022; 5:141-152. [PMID: 35343091 PMCID: PMC9043727 DOI: 10.1002/ame2.12223] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 01/07/2023] Open
Abstract
Animal models of human diseases play a critical role in medical research. Pigs are anatomically and physiologically more like humans than are small rodents such as mice, making pigs an attractive option for modeling human diseases. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, and various genetic diseases. We also discuss areas that need to be improved. Animal models of human diseases play a critical role in medical research. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, various genetic diseases and xenotransplantation.
Collapse
Affiliation(s)
- Naipeng Hou
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China
| | - Xuguang Du
- Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sen Wu
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
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: 0.5] [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
|
Kauser K, Warner KS, Anderson B, Keyes ED, Hayes RB, Kawamoto E, Perkins DH, Scott R, Isaacson J, Haberer B, Spaans A, Utecht R, Hauser H, Roberts AG, Greenberg M. Creating a Natural Vascular Scaffold by Photochemical Treatment of the Extracellular Matrix for Vascular Applications. Int J Mol Sci 2022; 23:ijms23020683. [PMID: 35054866 PMCID: PMC8775700 DOI: 10.3390/ijms23020683] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The development of bioscaffolds for cardiovascular medical applications, such as peripheral artery disease (PAD), remains to be a challenge for tissue engineering. PAD is an increasingly common and serious cardiovascular illness characterized by progressive atherosclerotic stenosis, resulting in decreased blood perfusion to the lower extremities. Percutaneous transluminal angioplasty and stent placement are routinely performed on these patients with suboptimal outcomes. Natural Vascular Scaffolding (NVS) is a novel treatment in the development for PAD, which offers an alternative to stenting by building on the natural structural constituents in the extracellular matrix (ECM) of the blood vessel wall. During NVS treatment, blood vessels are exposed to a photoactivatable small molecule (10-8-10 Dimer) delivered locally to the vessel wall via an angioplasty balloon. When activated with 450 nm wavelength light, this therapy induces the formation of covalent protein–protein crosslinks of the ECM proteins by a photochemical mechanism, creating a natural scaffold. This therapy has the potential to reduce the need for stent placement by maintaining a larger diameter post-angioplasty and minimizing elastic recoil. Experiments were conducted to elucidate the mechanism of action of NVS, including the molecular mechanism of light activation and the impact of NVS on the ECM.
Collapse
Affiliation(s)
- Katalin Kauser
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
- Correspondence: ; Tel.: +1-415-527-9892
| | - Kevin S. Warner
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Blake Anderson
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Edgar Dalles Keyes
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, UT 84112, USA; (E.D.K.); (A.G.R.)
| | - RB Hayes
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Eric Kawamoto
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - DH Perkins
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Robert Scott
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Jim Isaacson
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Barb Haberer
- Alumend, LLC, 4800 N. Career Avenue, Suite #108, Sioux Falls, SD 57107, USA; (B.H.); (A.S.); (R.U.)
| | - Ann Spaans
- Alumend, LLC, 4800 N. Career Avenue, Suite #108, Sioux Falls, SD 57107, USA; (B.H.); (A.S.); (R.U.)
| | - Ronald Utecht
- Alumend, LLC, 4800 N. Career Avenue, Suite #108, Sioux Falls, SD 57107, USA; (B.H.); (A.S.); (R.U.)
| | - Hank Hauser
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| | - Andrew George Roberts
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, UT 84112, USA; (E.D.K.); (A.G.R.)
| | - Myles Greenberg
- Alucent Biomedical Inc., 675 Arapeen Dr, Suite #102, Salt Lake City, UT 84108, USA; (K.S.W.); (B.A.); (R.H.); (E.K.); (D.P.); (R.S.); (J.I.); (H.H.); (M.G.)
| |
Collapse
|
16
|
Lunney JK, Van Goor A, Walker KE, Hailstock T, Franklin J, Dai C. Importance of the pig as a human biomedical model. Sci Transl Med 2021; 13:eabd5758. [PMID: 34818055 DOI: 10.1126/scitranslmed.abd5758] [Citation(s) in RCA: 268] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Angelica Van Goor
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Taylor Hailstock
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Jasmine Franklin
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Chaohui Dai
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA.,College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
17
|
Use of Agriculturally Important Animals as Models in Biomedical Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:315-333. [PMID: 34807449 DOI: 10.1007/978-3-030-85686-1_16] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Livestock have contributed significantly to advances in biomedicine and offer unique advantages over rodent models. The human is the ideal biomedical model; however, ethical reasons limit the testing of hypotheses and treatments in humans. Rodent models are frequently used as alternatives to humans due to size, low cost, and ease of genetic manipulation, and have contributed tremendously to our understanding of human health and disease. However, the use of rodents in translational research pose challenges for researchers due to physiological differences to humans. The use of livestock species as biomedical models can address these challenges as livestock have several similarities to human anatomy, physiology, genetics, and metabolism and their larger size permits collection of more frequent and often larger samples. Additionally, recent advances in genetics in livestock species allow for studies in genomics, proteomics, and metabolomics, which have the added benefit of applications to both humans in biomedical research and livestock in improving production. In this review, we provide an overview of scientific findings using livestock and benefits of each model to the livestock industry and to biomedical research.
Collapse
|
18
|
Animal Models of Neointimal Hyperplasia and Restenosis: Species-Specific Differences and Implications for Translational Research. JACC Basic Transl Sci 2021; 6:900-917. [PMID: 34869956 PMCID: PMC8617545 DOI: 10.1016/j.jacbts.2021.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/29/2022]
Abstract
Neointimal hyperplasia is the major factor contributing to restenosis after angioplasty procedures. Multiple animal models exist to study basic and translational aspects of restenosis formation. Animal models differ substantially, and species-specific differences have major impact on the pathophysiology of the model. Genetic, dietary, and mechanical interventions determine the translational potential of the animal model used and have to be considered when choosing the model.
The process of restenosis is based on the interplay of various mechanical and biological processes triggered by angioplasty-induced vascular trauma. Early arterial recoil, negative vascular remodeling, and neointimal formation therefore limit the long-term patency of interventional recanalization procedures. The most serious of these processes is neointimal hyperplasia, which can be traced back to 4 main mechanisms: endothelial damage and activation; monocyte accumulation in the subintimal space; fibroblast migration; and the transformation of vascular smooth muscle cells. A wide variety of animal models exists to investigate the underlying pathophysiology. Although mouse models, with their ease of genetic manipulation, enable cell- and molecular-focused fundamental research, and rats provide the opportunity to use stent and balloon models with high throughput, both rodents lack a lipid metabolism comparable to humans. Rabbits instead build a bridge to close the gap between basic and clinical research due to their human-like lipid metabolism, as well as their size being accessible for clinical angioplasty procedures. Every different combination of animal, dietary, and injury model has various advantages and disadvantages, and the decision for a proper model requires awareness of species-specific biological properties reaching from vessel morphology to distinct cellular and molecular features.
Collapse
Key Words
- Apo, apolipoprotein
- CETP, cholesteryl ester transferase protein
- ECM, extracellular matrix
- FGF, fibroblast growth factor
- HDL, high-density lipoprotein
- LDL, low-density lipoprotein
- LDLr, LDL receptor
- PDGF, platelet-derived growth factor
- TGF, transforming growth factor
- VLDL, very low-density lipoprotein
- VSMC, vascular smooth muscle cell
- angioplasty
- animal model
- neointimal hyperplasia
- restenosis
Collapse
|
19
|
Liu G, Lai P, Guo J, Wang Y, Xian X. Genetically-engineered hamster models: applications and perspective in dyslipidemia and atherosclerosis-related cardiovascular disease. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:92-110. [PMID: 37724074 PMCID: PMC10388752 DOI: 10.1515/mr-2021-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in both developed and developing countries, in which atherosclerosis triggered by dyslipidemia is the major pathological basis. Over the past 40 years, small rodent animals, such as mice, have been widely used for understanding of human atherosclerosis-related cardiovascular disease (ASCVD) with the advantages of low cost and ease of maintenance and manipulation. However, based on the concept of precision medicine and high demand of translational research, the applications of mouse models for human ASCVD study would be limited due to the natural differences in metabolic features between mice and humans even though they are still the most powerful tools in this research field, indicating that other species with biological similarity to humans need to be considered for studying ASCVD in future. With the development and breakthrough of novel gene editing technology, Syrian golden hamster, a small rodent animal replicating the metabolic characteristics of humans, has been genetically modified, suggesting that gene-targeted hamster models will provide new insights into the precision medicine and translational research of ASCVD. The purpose of this review was to summarize the genetically-modified hamster models with dyslipidemia to date, and their potential applications and perspective for ASCVD.
Collapse
Affiliation(s)
- George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Pingping Lai
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
20
|
Otake M, Kawaguchi H, Enya S, Kangawa A, Koga T, Matsuo K, Yamada S, Rahman MM, Miura N, Shibata M, Tanimoto A. High Pathological Reproducibility of Diet-induced Atherosclerosis in Microminipigs via Cloning Technology. In Vivo 2021; 35:2025-2033. [PMID: 34182477 DOI: 10.21873/invivo.12471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND/AIM The reproducibility of athero - sclerotic lesions was evaluated after the production of cloned-microminipigs and their offspring. MATERIALS AND METHODS Cloned-microminipig-parents were produced by microminipigsomatic cell nuclei. These parents were crossbred and delivered males (F1-offspring) were divided into two groups: normal chow diet (NcD)-fed and high-fat/high-cholesterol diet (HcD)-fed groups. One of the F1-offsprings was subjected to cloning, and delivered males (F1-clones) were fed with HcD. After 8 weeks, all animals were necropsied for patho - physiological studies compared to non-cloned-microminipigs. RESULTS HcD-fed F1-offspring and F1-clones, but not NcD-fed F1-offspring, exhibited increased serum lipid levels and systemic atherosclerosis, which were comparable to those of HcD-fed non-cloned-microminipigs. Homogeneity of variance analysis demonstrated that standard deviation values of serum lipoprotein and aortic atherosclerosis area from HcD-fed animals decreased in F1-offspring and F1-clones. CONCLUSION HcD-induced atherogenesis was highly reproducible in F1-offsprings and F1-clones, indicating that the atherosclerosis-prone genomic background was preserved in the cloned-microminipigs, which can be used for studies on human atherosclerosis and related diseases.
Collapse
Affiliation(s)
- Masayoshi Otake
- Swine and Poultry Department, Shizuoka Prefectural Research Institute of Animal Industry, Swine and Poultry Research Center, Kikugawa, Japan;
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Laboratory of Veterinary Pathology, School of Veterinary Medicine, Kitasato University, Towadashi, Japan
| | - Satoko Enya
- Swine and Poultry Department, Shizuoka Prefectural Research Institute of Animal Industry, Swine and Poultry Research Center, Kikugawa, Japan
| | - Akihisa Kangawa
- Swine and Poultry Department, Shizuoka Prefectural Research Institute of Animal Industry, Swine and Poultry Research Center, Kikugawa, Japan
| | - Tadashi Koga
- Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Japan
| | - Kei Matsuo
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Sohsuke Yamada
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Kahoku, Japan
| | - Md Mahfuzur Rahman
- Veterinary Teaching Hospital, Joint faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Masatoshi Shibata
- Swine and Poultry Department, Shizuoka Prefectural Research Institute of Animal Industry, Swine and Poultry Research Center, Kikugawa, Japan
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan;
| |
Collapse
|
21
|
Han Y, Zhang Y, Liu S, Chen G, Cao L, Xin Y. Association of LDLR rs1433099 with the Risk of NAFLD and CVD in Chinese Han Population. J Clin Transl Hepatol 2021; 9:203-209. [PMID: 34007802 PMCID: PMC8111099 DOI: 10.14218/jcth.2020.00163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/01/2021] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS Recent genome-wide association studies have shown that low-density lipoprotein receptor (LDLR) rs1433099 polymorphism is associated with cardiovascular disease (CVD) risk in many countries. However, the association of LDLR rs1433099 with CVD in China has not been reported yet. There are no studies on LDLR rs1433099 and non-alcoholic fatty liver disease (NAFLD) as well. The purpose of this study was to investigate whether LDLR rs1433099 is related to CVD or NAFLD in the Chinese population. METHODS LDLR rs1433099 polymorphism was genotyped in 507 individuals, including 140 healthy controls, 79 NAFLD patients, 185 CVD patients, and 103 patients with NAFLD combined with CVD. The expression of LDLR was tested by the sequence detection system, and clinical parameters were assessed by biochemical tests and physical examination. RESULTS The genotype distribution of LDLR rs1433099 was not statistically different among the NAFLD group, the CVD group, the combined group, and the healthy control group (p>0.05). There was no significant correlation of LDLR rs1433099 genotypic distribution or allele frequency and the risk of NAFLD, CVD or NAFLD combined with CVD (p>0.05). In the CVD group, T allele carriers had higher alkaline phosphatase and gamma-glutamyl transpeptidase than non-carriers (p<0.05). CONCLUSIONS Our study demonstrated that the LDLR rs1433099 polymorphism is not a risk factor of NAFLD. The LDLR rs1433099 polymorphism may increase the risk of CVD through a mechanism involving alkaline phosphatase and gamma-glutamyl transpeptidase.
Collapse
Affiliation(s)
- Yi Han
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, Shandong, China
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongshuo Zhang
- Administrative Management Office, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shousheng Liu
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Guangxia Chen
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linlin Cao
- Department of Gastroenterology, The First People’s Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongning Xin
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, Shandong, China
- Correspondence to: Yongning Xin, Department of Infectious Disease, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong 266011, China. ORCID: http://orcid.org/0000-0002-3692-7655. Tel: +86-532-82789463, Fax: +86-532-85968434, E-mail:
| |
Collapse
|
22
|
Iop L. Toward the Effective Bioengineering of a Pathological Tissue for Cardiovascular Disease Modeling: Old Strategies and New Frontiers for Prevention, Diagnosis, and Therapy. Front Cardiovasc Med 2021; 7:591583. [PMID: 33748193 PMCID: PMC7969521 DOI: 10.3389/fcvm.2020.591583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVDs) still represent the primary cause of mortality worldwide. Preclinical modeling by recapitulating human pathophysiology is fundamental to advance the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and treatment. In silico, in vivo, and in vitro models have been applied to dissect many cardiovascular pathologies. Computational and bioinformatic simulations allow developing algorithmic disease models considering all known variables and severity degrees of disease. In vivo studies based on small or large animals have a long tradition and largely contribute to the current treatment and management of CVDs. In vitro investigation with two-dimensional cell culture demonstrates its suitability to analyze the behavior of single, diseased cellular types. The introduction of induced pluripotent stem cell technology and the application of bioengineering principles raised the bar toward in vitro three-dimensional modeling by enabling the development of pathological tissue equivalents. This review article intends to describe the advantages and disadvantages of past and present modeling approaches applied to provide insights on some of the most relevant congenital and acquired CVDs, such as rhythm disturbances, bicuspid aortic valve, cardiac infections and autoimmunity, cardiovascular fibrosis, atherosclerosis, and calcific aortic valve stenosis.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences, and Public Health, University of Padua Medical School, Padua, Italy
| |
Collapse
|
23
|
Cheng AA, Li W, Walker TM, Silvers C, Arendt LM, Hernandez LL. Investigating the complex interplay between genotype and high-fat-diet feeding in the lactating mammary gland using the Tph1 and Ldlr knockout models. Am J Physiol Endocrinol Metab 2021; 320:E438-E452. [PMID: 33427054 PMCID: PMC7988787 DOI: 10.1152/ajpendo.00456.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022]
Abstract
Obesity is a prevailing problem across the globe. Women who are obese have difficulty initiating and sustaining lactation. However, the impact of genetics and diet on breastfeeding outcomes is understudied. Here we explore the effect of diet and genotype on lactation. We utilized the low-density lipoprotein receptor (Ldlr-KO) transgenic mouse model as an obesity and hypercholesterolemia model. Additionally, we used the tryptophan hydroxylase 1 (Tph1-KO) mouse, recently identified as a potential anti-obesogenic model, to investigate if addition of Tph1-KO could ameliorate negative effects of obesity in Ldlr-KO mice. We created a novel transgenic mouse line by combining the Ldlr and Tph1 [double knockout (DKO)] mice to study the interaction between the two genotypes. Female mice were fed a low-fat diet (LFD; 10% fat) or high-fat diet (HFD; 60% fat) from 3 wk of age through early [lactation day 3 (L3)] or peak lactation [lactation day 11 (L11)]. After 4 wk of consuming either LFD or HFD, female mice were bred. On L2 and L10, dams were milked to investigate the effect of diet and genotype on milk composition. Dams were euthanized on L3 or L11. There was no impact of diet or genotype on milk protein or triglycerides (TGs) on L2; however, by L10, Ldlr-KO and DKO dams had increased TG levels in milk. RNA-sequencing of L11 mammary glands demonstrated Ldlr-KO dams fed HFD displayed enrichment of genes involved in immune system pathways. Interestingly, the DKO may alter vesicle budding and biogenesis during lactation. We also quantified macrophages by immunostaining for F4/80+ cells at L3 and L11. Diet played a significant role on L3 (P = 0.013), but genotype played a role at L11 (P < 0.0001) on numbers of F4/80+ cells. Thus the impact of diet and genotype on lactation differs depending on stage of lactation, illustrating complexities of understanding the intersection of these parameters.NEW & NOTEWORTHY We have created a novel mouse model that is focused on understanding the intersection of diet and genotype on mammary gland function during lactation.
Collapse
Affiliation(s)
- Adrienne A Cheng
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, Wisconsin
| | - Wenli Li
- US Department of Agriculture-Dairy Forage, Madison, Wisconsin
| | - Teresa M Walker
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, Wisconsin
| | - Caylee Silvers
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Lisa M Arendt
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Laura L Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
24
|
Koppes EA, Redel BK, Johnson MA, Skvorak KJ, Ghaloul-Gonzalez L, Yates ME, Lewis DW, Gollin SM, Wu YL, Christ SE, Yerle M, Leshinski A, Spate LD, Benne JA, Murphy SL, Samuel MS, Walters EM, Hansen SA, Wells KD, Lichter-Konecki U, Wagner RA, Newsome JT, Dobrowolski SF, Vockley J, Prather RS, Nicholls RD. A porcine model of phenylketonuria generated by CRISPR/Cas9 genome editing. JCI Insight 2020; 5:141523. [PMID: 33055427 PMCID: PMC7605535 DOI: 10.1172/jci.insight.141523] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Phenylalanine hydroxylase-deficient (PAH-deficient) phenylketonuria (PKU) results in systemic hyperphenylalaninemia, leading to neurotoxicity with severe developmental disabilities. Dietary phenylalanine (Phe) restriction prevents the most deleterious effects of hyperphenylalaninemia, but adherence to diet is poor in adult and adolescent patients, resulting in characteristic neurobehavioral phenotypes. Thus, an urgent need exists for new treatments. Additionally, rodent models of PKU do not adequately reflect neurocognitive phenotypes, and thus there is a need for improved animal models. To this end, we have developed PAH-null pigs. After selection of optimal CRISPR/Cas9 genome-editing reagents by using an in vitro cell model, zygote injection of 2 sgRNAs and Cas9 mRNA demonstrated deletions in preimplantation embryos, with embryo transfer to a surrogate leading to 2 founder animals. One pig was heterozygous for a PAH exon 6 deletion allele, while the other was compound heterozygous for deletions of exon 6 and of exons 6-7. The affected pig exhibited hyperphenylalaninemia (2000-5000 μM) that was treatable by dietary Phe restriction, consistent with classical PKU, along with juvenile growth retardation, hypopigmentation, ventriculomegaly, and decreased brain gray matter volume. In conclusion, we have established a large-animal preclinical model of PKU to investigate pathophysiology and to assess new therapeutic interventions.
Collapse
Affiliation(s)
- Erik A. Koppes
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bethany K. Redel
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Marie A. Johnson
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kristen J. Skvorak
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lina Ghaloul-Gonzalez
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Megan E. Yates
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dale W. Lewis
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Susanne M. Gollin
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Yijen L. Wu
- Department of Developmental Biology, University of Pittsburgh, and UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shawn E. Christ
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, USA
| | - Martine Yerle
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet-Tolosan, France
| | - Angela Leshinski
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lee D. Spate
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Joshua A. Benne
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Stephanie L. Murphy
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Melissa S. Samuel
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Eric M. Walters
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Sarah A. Hansen
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Kevin D. Wells
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Uta Lichter-Konecki
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert A. Wagner
- Division of Laboratory Animal Resources, Office of Research, Health Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph T. Newsome
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Laboratory Animal Resources, Office of Research, Health Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven F. Dobrowolski
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jerry Vockley
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Randall S. Prather
- Division ofAnimal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, Missouri, USA
- National Swine Research and Resource Center (NSRRC), College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Robert D. Nicholls
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, and Universityof Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Hryhorowicz M, Lipiński D, Hryhorowicz S, Nowak-Terpiłowska A, Ryczek N, Zeyland J. Application of Genetically Engineered Pigs in Biomedical Research. Genes (Basel) 2020; 11:genes11060670. [PMID: 32575461 PMCID: PMC7349405 DOI: 10.3390/genes11060670] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Progress in genetic engineering over the past few decades has made it possible to develop methods that have led to the production of transgenic animals. The development of transgenesis has created new directions in research and possibilities for its practical application. Generating transgenic animal species is not only aimed towards accelerating traditional breeding programs and improving animal health and the quality of animal products for consumption but can also be used in biomedicine. Animal studies are conducted to develop models used in gene function and regulation research and the genetic determinants of certain human diseases. Another direction of research, described in this review, focuses on the use of transgenic animals as a source of high-quality biopharmaceuticals, such as recombinant proteins. The further aspect discussed is the use of genetically modified animals as a source of cells, tissues, and organs for transplantation into human recipients, i.e., xenotransplantation. Numerous studies have shown that the pig (Sus scrofa domestica) is the most suitable species both as a research model for human diseases and as an optimal organ donor for xenotransplantation. Short pregnancy, short generation interval, and high litter size make the production of transgenic pigs less time-consuming in comparison with other livestock species This review describes genetically modified pigs used for biomedical research and the future challenges and perspectives for the use of the swine animal models.
Collapse
Affiliation(s)
- Magdalena Hryhorowicz
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
- Correspondence:
| | - Daniel Lipiński
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Szymon Hryhorowicz
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland;
| | - Agnieszka Nowak-Terpiłowska
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Natalia Ryczek
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Joanna Zeyland
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| |
Collapse
|
26
|
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: 1.0] [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
|
27
|
Abstract
Atherosclerosis is a chronic inflammatory vascular disease and the predominant cause of heart attack and ischemic stroke. Despite the well-known sexual dimorphism in the incidence and complications of atherosclerosis, there are relatively limited data in the clinical and preclinical literature to rigorously address mechanisms underlying sex as a biological variable in atherosclerosis. In multiple histological and imaging studies, overall plaque burden and markers of inflammation appear to be greater in men than women and are predictive of cardiovascular events. However, while younger women are relatively protected from cardiovascular disease, by the seventh decade, the incidence of myocardial infarction in women ultimately surpasses that of men, suggesting an interaction between sex and age. Most preclinical studies in animal atherosclerosis models do not examine both sexes, and even in those that do, well-powered direct statistical comparisons for sex as an independent variable remain rare. This article reviews the available data. Overall, male animals appear to have more inflamed yet smaller plaques compared to female animals. Plaque inflammation is often used as a surrogate end point for plaque vulnerability in animals. The available data support the notion that rather than plaque size, plaque inflammation may be more relevant in assessing sex-specific mechanisms since the findings correlate with the sex difference in ischemic events and mortality and thus may be more reflective of the human condition. Overall, the number of preclinical studies directly comparing plaque inflammation between the sexes is extremely limited relative to the vast literature exploring atherosclerosis mechanisms. Failure to include both sexes and to address age in mechanistic atherosclerosis studies are missed opportunities to uncover underlying sex-specific mechanisms. Understanding the mechanisms driving sex as a biological variable in atherosclerotic disease is critical to future precision medicine strategies to mitigate what is still the leading cause of death of men and women worldwide.
Collapse
Affiliation(s)
- Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Joshua A. Beckman
- Cardiovascular Division, Vanderbilt University Medical Center, Nashville, TN
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| |
Collapse
|
28
|
Yamashita S, Kogasaka Y, Hiradate Y, Tanemura K, Sendai Y. Suppression of mosaic mutation by co-delivery of CRISPR associated protein 9 and three-prime repair exonuclease 2 into porcine zygotes via electroporation. J Reprod Dev 2020; 66:41-48. [PMID: 31761839 PMCID: PMC7040215 DOI: 10.1262/jrd.2019-088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Gene-modified animals, including pigs, can be generated efficiently by introducing CRISPR associated protein 9 (CRISPR/Cas9) into zygotes. However, in many cases, these zygotes tend to become mosaic mutants with various different mutant cell types, making it difficult to analyze the phenotype of gene-modified founder animals. To reduce the mosaic mutations, we introduced three-prime repair exonuclease 2 (Trex2), an exonuclease that improves gene editing efficiency, into porcine zygotes along with CRISPR/Cas9 via electroporation. Although the rate of porcine blastocyst formation decreased due to electroporation (25.9 ± 4.6% vs. 41.2 ± 2.0%), co-delivery of murine Trex2 (mTrex2) mRNA with CRISPR/Cas9 did not affect it any further (25.9 ± 4.6% vs. 31.0 ± 4.6%). In addition, there was no significant difference in the diameter of blastocysts carrying CRISPR/Cas9 (164.7 ± 10.2 μm), and those with CRISPR/Cas9 + mTrex2 (151.9 ± 5.1 μm) as compared to those from the control group (178.9 ± 9.0 μm). These results revealed that mTrex2 did not affect the development of pre-implantation embryo. We also found bi-allelic, as well as mono-allelic, non-mosaic homozygous mutations in the blastocysts. Most importantly, co-delivery of mTrex2 mRNA with CRISPR/Cas9 increased non-mosaic mutant blastocysts (29.3 ± 4.5%) and reduced mosaic mutant blastocysts (70.7 ± 4.5%) as compared to CRISPR/Cas9 alone (5.6 ± 6.4% and 92.6 ± 8.6%, respectively). These data suggest that the co-delivery of CRISPR/Cas9 and mTrex2 is a useful method to suppress mosaic mutation.
Collapse
Affiliation(s)
- Shiro Yamashita
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Yuhei Kogasaka
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
| | - Yuuki Hiradate
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Yutaka Sendai
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
| |
Collapse
|
29
|
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: 11.5] [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
|
30
|
Kalla D, Kind A, Schnieke A. Genetically Engineered Pigs to Study Cancer. Int J Mol Sci 2020; 21:E488. [PMID: 31940967 PMCID: PMC7013672 DOI: 10.3390/ijms21020488] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent decades have seen groundbreaking advances in cancer research. Genetically engineered animal models, mainly in mice, have contributed to a better understanding of the underlying mechanisms involved in cancer. However, mice are not ideal for translating basic research into studies closer to the clinic. There is a need for complementary information provided by non-rodent species. Pigs are well suited for translational biomedical research as they share many similarities with humans such as body and organ size, aspects of anatomy, physiology and pathophysiology and can provide valuable means of developing and testing novel diagnostic and therapeutic procedures. Porcine oncology is a new field, but it is clear that replication of key oncogenic mutation in pigs can usefully mimic several human cancers. This review briefly outlines the technology used to generate genetically modified pigs, provides an overview of existing cancer models, their applications and how the field may develop in the near future.
Collapse
Affiliation(s)
| | | | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany; (D.K.); (A.K.)
| |
Collapse
|
31
|
Suva LJ, Westhusin ME, Long CR, Gaddy D. Engineering bone phenotypes in domestic animals: Unique resources for enhancing musculoskeletal research. Bone 2020; 130:115119. [PMID: 31712131 PMCID: PMC8805042 DOI: 10.1016/j.bone.2019.115119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College Station, TX, 77843, United States.
| | - Mark E Westhusin
- Department of Veterinary Physiology and Pharmacology, College Station, TX, 77843, United States
| | - Charles R Long
- Department of Veterinary Physiology and Pharmacology, College Station, TX, 77843, United States
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences Texas A&M University, College Station, TX 77843, United States
| |
Collapse
|
32
|
Kee PH, Moody MR, Huang SL, Kim H, Yin X, Peng T, Laing ST, Klegerman ME, Rahbar MH, Vela D, Genstler C, Haworth KJ, Holland CK, McPherson DD. Stabilizing Peri-Stent Restenosis Using a Novel Therapeutic Carrier. JACC Basic Transl Sci 2020; 5:1-11. [PMID: 32043017 PMCID: PMC7000871 DOI: 10.1016/j.jacbts.2019.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
Late in-stent restenosis remains a significant problem. Bare-metal stents were implanted into peripheral arteries in miniature swine, followed by direct intra-arterial infusion of nitric oxide-loaded echogenic liposomes (ELIPs) and anti-intercellular adhesion molecule-1 conjugated ELIPs loaded with pioglitazone exposed to an endovascular catheter with an ultrasonic core. Ultrasound-facilitated delivery of ELIP formulations into stented peripheral arteries attenuated neointimal growth. Local atheroma-targeted, ultrasound-triggered delivery of nitric oxide and pioglitazone, an anti-inflammatory peroxisome proliferator-activated receptor-γ agonist, into stented arteries has the potential to stabilize stent-induced neointimal growth and obviate the need for long-term antiplatelet therapy.
Collapse
Affiliation(s)
- Patrick H. Kee
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Melanie R. Moody
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Shao-Ling Huang
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Hyunggun Kim
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
- Department of Bio-Mechatronic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Xing Yin
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Tao Peng
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Susan T. Laing
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Melvin E. Klegerman
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Mohammad H. Rahbar
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Deborah Vela
- Department of Pathology, Texas Heart Institute, Houston, Texas
| | | | - Kevin J. Haworth
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Christy K. Holland
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - David D. McPherson
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
33
|
Li DY, Busch A, Jin H, Chernogubova E, Pelisek J, Karlsson J, Sennblad B, Liu S, Lao S, Hofmann P, Bäcklund A, Eken SM, Roy J, Eriksson P, Dacken B, Ramanujam D, Dueck A, Engelhardt S, Boon RA, Eckstein HH, Spin JM, Tsao PS, Maegdefessel L. H19 Induces Abdominal Aortic Aneurysm Development and Progression. Circulation 2019; 138:1551-1568. [PMID: 29669788 DOI: 10.1161/circulationaha.117.032184] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Long noncoding RNAs have emerged as critical molecular regulators in various biological processes and diseases. Here we sought to identify and functionally characterize long noncoding RNAs as potential mediators in abdominal aortic aneurysm development. METHODS We profiled RNA transcript expression in 2 murine abdominal aortic aneurysm models, Angiotensin II (ANGII) infusion in apolipoprotein E-deficient ( ApoE-/-) mice (n=8) and porcine pancreatic elastase instillation in C57BL/6 wild-type mice (n=12). The long noncoding RNA H19 was identified as 1 of the most highly upregulated transcripts in both mouse aneurysm models compared with sham-operated controls. This was confirmed by quantitative reverse transcription-polymerase chain reaction and in situ hybridization. RESULTS Experimental knock-down of H19, utilizing site-specific antisense oligonucleotides (LNA-GapmeRs) in vivo, significantly limited aneurysm growth in both models. Upregulated H19 correlated with smooth muscle cell (SMC) content and SMC apoptosis in progressing aneurysms. Importantly, a similar pattern could be observed in human abdominal aortic aneurysm tissue samples, and in a novel preclinical LDLR-/- (low-density lipoprotein receptor) Yucatan mini-pig aneurysm model. In vitro knock-down of H19 markedly decreased apoptotic rates of cultured human aortic SMCs, whereas overexpression of H19 had the opposite effect. Notably, H19-dependent apoptosis mechanisms in SMCs appeared to be independent of miR-675, which is embedded in the first exon of the H19 gene. A customized transcription factor array identified hypoxia-inducible factor 1α as the main downstream effector. Increased SMC apoptosis was associated with cytoplasmic interaction between H19 and hypoxia-inducible factor 1α and sequential p53 stabilization. Additionally, H19 induced transcription of hypoxia-inducible factor 1α via recruiting the transcription factor specificity protein 1 to the promoter region. CONCLUSIONS The long noncoding RNA H19 is a novel regulator of SMC survival in abdominal aortic aneurysm development and progression. Inhibition of H19 expression might serve as a novel molecular therapeutic target for aortic aneurysm disease.
Collapse
Affiliation(s)
- Daniel Y Li
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Albert Busch
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Hong Jin
- Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| | - Ekaterina Chernogubova
- Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Joakim Karlsson
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden (J.K.)
| | - Bengt Sennblad
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Sweden (B.S.)
| | - Shengliang Liu
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Shen Lao
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Patrick Hofmann
- Institute of Cardiovascular Regeneration, University Hospital Frankfurt, and German Center for Cardiovascular Research (DZHK), partner site Rhein-Main, Frankfurt, Germany (P.H., R.A.B.)
| | - Alexandra Bäcklund
- Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| | - Suzanne M Eken
- Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| | - Joy Roy
- Department of Molecular Medicine and Surgery (J.R.), Karolinska Institutet, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| | | | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology (D.R., A.D., S.E.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Anne Dueck
- Institute of Pharmacology and Toxicology (D.R., A.D., S.E.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology (D.R., A.D., S.E.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Reinier A Boon
- Institute of Cardiovascular Regeneration, University Hospital Frankfurt, and German Center for Cardiovascular Research (DZHK), partner site Rhein-Main, Frankfurt, Germany (P.H., R.A.B.)
| | - Hans-Henning Eckstein
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University, CA (J.M.S., P.S.T.)
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University, CA (J.M.S., P.S.T.)
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar (D.Y.L., A. Busch, J.P., S.L., H.-H.E., L.M.), Technical University Munich, and German Center for Cardiovascular Research (DZHK), partner site Munich, Germany.,Department of Medicine (H.J., E.C., A. Bäcklund; S.M.E., P.E., L.M.), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
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: 19] [Impact Index Per Article: 3.8] [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
|
35
|
Mushenkova NV, Summerhill VI, Silaeva YY, Deykin AV, Orekhov AN. Modelling of atherosclerosis in genetically modified animals. Am J Transl Res 2019; 11:4614-4633. [PMID: 31497187 PMCID: PMC6731422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Atherosclerosis is a lipid-driven, chronic inflammatory disease that leads to plaque formation at specific sites of the arterial tree. Being the common cause of many cardiovascular disorders, atherosclerosis makes a tremendous impact on morbidity and mortality rates of cardiovascular diseases (CVDs) in countries with higher income. Animal models of atherosclerosis are utilized as useful tools for studying the aetiology, pathogenesis and complications of atherosclerosis, thus, providing a valuable platform for the efficacy testing of different pharmacological therapies and validation of imaging techniques. To date, a large variety of models is available. Pathophysiological changes can be induced in animals by either an atherogenic diet or genetic manipulations. The discussion of advantages and disadvantages of some murine, rabbit and porcine genetic models currently available for the atherosclerosis research is the scope of the following review.
Collapse
Affiliation(s)
| | - Volha I Summerhill
- Institute for Atherosclerosis Research, Skolkovo Innovative CentreMoscow 121609, Russia
| | - Yulia Yu Silaeva
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexey V Deykin
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative CentreMoscow 121609, Russia
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| |
Collapse
|
36
|
Dietary Cholesterol Is Highly Associated with Severity of Hyperlipidemia and Atherosclerotic Lesions in Heterozygous LDLR-Deficient Hamsters. Int J Mol Sci 2019; 20:ijms20143515. [PMID: 31323736 PMCID: PMC6678973 DOI: 10.3390/ijms20143515] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/14/2019] [Accepted: 07/15/2019] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Familial hypercholesterolemia (FH) is a dominant inherited disease caused mainly by low-density lipoprotein receptor (LDLR) gene mutations. To different extents, both heterozygous and homozygous FH patients develop premature coronary heart disease (CHD). However, most of the experimental animal models with LDLR deficiency could not fully recapitulate FH because they develop hyperlipidemia and atherosclerosis only in homozygous, but not in heterozygous, form. In the current study, we investigated the responsiveness of the LDLR+/- hamster to dietary cholesterol and whether plasma cholesterol levels were positively associated with the severity of atherosclerosis. Approach and Methods: wild type WT and LDLR+/- hamsters were fed a high fat diet with different cholesterol contents (HCHF) for 12 or 16 weeks. Plasma lipids, (apo)lipoproteins, and atherosclerosis in both the aorta and coronary arteries were analyzed. After a HCHF diet challenge, the levels of total cholesterol (TC) in WT and LDLR+/- hamsters were significantly elevated, but the latter showed a more pronounced lipoprotein profile, with higher cholesterol levels that were positively correlated with dietary cholesterol contents. The LDLR+/- hamsters also showed accelerated atherosclerotic lesions in the aorta and coronary arteries, whereas only mild aortic lesions were observed in WT hamsters. CONCLUSIONS Our findings demonstrate that, unlike other rodent animals, the levels of plasma cholesterol in hamsters can be significantly modulated by the intervention of dietary cholesterol, which were closely associated with severity of atherosclerosis in LDLR+/- hamsters, suggesting that the LDLR+/- hamster is an ideal animal model for FH and has great potential in the study of FH and atherosclerosis-related CHD.
Collapse
|
37
|
He K, Wang J, Shi H, Yu Q, Zhang X, Guo M, Sun H, Lin X, Wu Y, Wang L, Wang Y, Xian X, Liu G. An interspecies study of lipid profiles and atherosclerosis in familial hypercholesterolemia animal models with low-density lipoprotein receptor deficiency. Am J Transl Res 2019; 11:3116-3127. [PMID: 31217881 PMCID: PMC6556657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/25/2019] [Indexed: 06/09/2023]
Abstract
Small rodents, especially mice and rats, have been widely used in atherosclerosis studies even though humans exhibit completely different lipoprotein metabolism and atherosclerotic characteristics. Until recently, various rodent models of human familial hypercholesterolemia (FH) have been created, including mice, rats, and golden Syrian hamsters. Although hamsters reportedly possess metabolic features similar to humans, there is no systematic characterization of the properties of circulating lipids and atherosclerotic lesions in these rodent models. We used three FH animal species (mice, rats, and hamsters) with low-density lipoprotein receptor (Ldlr) deficiency to fully assess lipoprotein metabolism and atherosclerotic characteristics. Compared to chow diet-fed mice and rats, Ldlr knockout (KO) hamsters showed increased cholesterols in LDL fractions similar to human FH patients. Upon 12-week high-cholesterol/high-fat diet feeding, both heterozygous and homozygous Ldlr KO hamsters displayed hyperlipidemic phenotypes, whereas only homozygous Ldlr KO mice and rats showed only moderate increases in plasma lipid levels. Moreover, rats were resistant to diet-induced atherosclerosis compared to mice, and hamsters showed more atherosclerotic lesions in the aortas and coronary arteries. Further morphological study revealed that only hamsters developed atherosclerosis in the abdominal segments, which is highly similar to FH patients. This unique animal model will provide insight into the translational study of human atherosclerosis and could be useful for developing novel treatments for FH patients.
Collapse
Affiliation(s)
- Kunxiang He
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Jinjie Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Haozhe Shi
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Qiongyang Yu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Xin Zhang
- Hebei Invivo Biotech CoShijiazhuang, China
| | - Mengmeng Guo
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Huijun Sun
- College of Pharmacy, Dalian Medical UniversityDalian 116044, China
| | - Xiao Lin
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Yue Wu
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing An Zhen Hospital Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing 100029, China
| | - Luya Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing An Zhen Hospital Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing 100029, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| | - Xunde Xian
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas 75390, Texas, USA
| | - George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking UniversityBeijing 100191, China
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Bempedoic acid has emerged as a potent inhibitor of ATP-citrate lyase (ACLY), a target for the reduction of LDL cholesterol (LDL-C). We review the impact of bempedoic acid treatment on lipoprotein metabolism and atherosclerosis in preclinical models and patients with hypercholesterolemia. RECENT FINDINGS The liver-specific activation of bempedoic acid inhibits ACLY, a key enzyme linking glucose catabolism to lipogenesis by catalyzing the formation of acetyl-CoA from mitochondrial-derived citrate for de novo synthesis of fatty acids and cholesterol. Adenosine monophosphate-activated protein kinase activation by bempedoic acid is not required for its lipid-regulating effects in vivo. Mendelian randomization of large human study cohorts has validated ACLY inhibition as a target for LDL-C lowering and atheroprotection. In rodents, bempedoic acid decreases plasma cholesterol and triglycerides, and prevents hepatic steatosis. In apolipoprotein E-deficient (Apoe) mice, LDL receptor-deficient (Ldlr) mice and LDLR-deficient miniature pigs, bempedoic acid reduces LDL-C and attenuates atherosclerosis. LDLR expression and activity are increased in primary human hepatocytes and in Apoe mouse liver treated with bempedoic acid suggesting a mechanism for LDL-C lowering, although additional pathways are likely involved. Phase 2 and 3 clinical trials revealed that bempedoic acid effectively lowers LDL-C as monotherapy, combined with ezetimibe, added to statin therapy and in statin-intolerant hypercholesterolemic patients. Treatment does not affect plasma concentrations of triglyceride or other lipoproteins. SUMMARY The LDL-C-lowering and attenuated atherosclerosis in animal models and reduced LDL-C in hypercholesterolemic patients has validated ACLY inhibition as a therapeutic strategy. Positive results from phase 3 long-term cardiovascular outcome trials in high-risk patients are required for bempedoic acid to be approved for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Amy C Burke
- Department of Biochemistry
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - Dawn E Telford
- Department of Medicine
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - Murray W Huff
- Department of Biochemistry
- Department of Medicine
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
39
|
Kinder HA, Baker EW, West FD. The pig as a preclinical traumatic brain injury model: current models, functional outcome measures, and translational detection strategies. Neural Regen Res 2019; 14:413-424. [PMID: 30539807 PMCID: PMC6334610 DOI: 10.4103/1673-5374.245334] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a major contributor of long-term disability and a leading cause of death worldwide. A series of secondary injury cascades can contribute to cell death, tissue loss, and ultimately to the development of functional impairments. However, there are currently no effective therapeutic interventions that improve brain outcomes following TBI. As a result, a number of experimental TBI models have been developed to recapitulate TBI injury mechanisms and to test the efficacy of potential therapeutics. The pig model has recently come to the forefront as the pig brain is closer in size, structure, and composition to the human brain compared to traditional rodent models, making it an ideal large animal model to study TBI pathophysiology and functional outcomes. This review will focus on the shared characteristics between humans and pigs that make them ideal for modeling TBI and will review the three most common pig TBI models-the diffuse axonal injury, the controlled cortical impact, and the fluid percussion models. It will also review current advances in functional outcome assessment measures and other non-invasive, translational TBI detection and measurement tools like biomarker analysis and magnetic resonance imaging. The use of pigs as TBI models and the continued development and improvement of translational assessment modalities have made significant contributions to unraveling the complex cascade of TBI sequela and provide an important means to study potential clinically relevant therapeutic interventions.
Collapse
Affiliation(s)
- Holly A Kinder
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Emily W Baker
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Franklin D West
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| |
Collapse
|
40
|
Fang B, Ren X, Wang Y, Li Z, Zhao L, Zhang M, Li C, Zhang Z, Chen L, Li X, Liu J, Xiong Q, Zhang L, Jin Y, Liu X, Li L, Wei H, Yang H, Li R, Dai Y. Apolipoprotein E deficiency accelerates atherosclerosis development in miniature pigs. Dis Model Mech 2018; 11:dmm036632. [PMID: 30305304 PMCID: PMC6215431 DOI: 10.1242/dmm.036632] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/28/2018] [Indexed: 12/26/2022] Open
Abstract
Miniature pigs have advantages over rodents in modeling atherosclerosis because their cardiovascular system and physiology are similar to that of humans. Apolipoprotein E (ApoE) deficiency has long been implicated in cardiovascular disease in humans. To establish an improved large animal model of familial hypercholesterolemia and atherosclerosis, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 system (CRISPR/Cas9) was used to disrupt the ApoE gene in Bama miniature pigs. Biallelic-modified ApoE pigs with in-frame mutations (ApoEm/m ) and frameshift mutations (ApoE-/- ) were simultaneously produced. ApoE-/- pigs exhibited moderately increased plasma cholesterol levels when fed with a regular chow diet, but displayed severe hypercholesterolemia and spontaneously developed human-like atherosclerotic lesions in the aorta and coronary arteries after feeding on a high-fat and high-cholesterol (HFHC) diet for 6 months. Thus, these ApoE-/- pigs could be valuable large animal models for providing further insight into translational studies of atherosclerosis.
Collapse
Affiliation(s)
- Bin Fang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xueyang Ren
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Ze Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Lihua Zhao
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Manling Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Chu Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Zhengwei Zhang
- Huai'an First Hospital Affiliated to Nanjing Medical University, Department of Pathology, Huai'an 223300, China
| | - Lei Chen
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoxue Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Xiong
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Lining Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Yong Jin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xiaorui Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Lin Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Haiyuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| |
Collapse
|
41
|
Spontaneous severe hypercholesterolemia and atherosclerosis lesions in rabbits with deficiency of low-density lipoprotein receptor (LDLR) on exon 7. EBioMedicine 2018; 36:29-38. [PMID: 30243490 PMCID: PMC6197696 DOI: 10.1016/j.ebiom.2018.09.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/02/2018] [Accepted: 09/12/2018] [Indexed: 11/20/2022] Open
Abstract
Rabbits (Oryctolagus cuniculus) have been the very frequently used as animal models in the study of human lipid metabolism and atherosclerosis, because they have similar lipoprotein metabolism to humans. Most of hyperlipidemia and atherosclerosis rabbit models are produced by feeding rabbits a high-cholesterol diet. Gene editing or knockout (KO) offered another means of producing rabbit models for study of the metabolism of lipids and lipoproteins. Even so, apolipoprotein (Apo)E KO rabbits must be fed a high-cholesterol diet to induce hyperlipidemia. In this study, we used the CRISPR/Cas9 system anchored exon 7 of low-density lipoprotein receptor (LDLR) in an attempt to generate KO rabbits. We designed two sgRNA sequences located in E7:g.7055-7074 and E7:g.7102-7124 of rabbit LDLR gene, respectively. Seven LDLR-KO founder rabbits were generated, and all of them contained biallelic modifications. Various mutational LDLR amino acid sequences of the 7 founder rabbits were subjected to tertiary structure modeling with SWISS-MODEL, and results showed that the structure of EGF-A domain of each protein differs from the wild-type. All the founder rabbits spontaneously developed hypercholesterolemia and atherosclerosis on a normal chow (NC) diet. Analysis of their plasma lipids and lipoproteins at the age of 12 weeks revealed that all these KO rabbits exhibited markedly increased levels of plasma TC (the highest of which was 1013.15 mg/dl, 20-fold higher than wild-type rabbits), LDL-C (the highest of which was 730.00 mg/dl, 35-fold higher than wild-type rabbits) and TG accompanied by reduced HDL-C levels. Pathological examinations of a founder rabbit showed prominent aortic atherosclerosis lesions and coronary artery atherosclerosis.In conclusion, we have reported the generation LDLR-KO rabbit model for the study of spontaneous hypercholesterolemia and atherosclerosis on a NC diet. The LDLR-KO rabbits should be a useful rabbit model of human familial hypercholesterolemia (FH) for the simulations of human primary hypercholesterolemia and such models would allow more exact research into cardio-cerebrovascular disease.
Collapse
|
42
|
Yang H, Wu Z. Genome Editing of Pigs for Agriculture and Biomedicine. Front Genet 2018; 9:360. [PMID: 30233645 PMCID: PMC6131568 DOI: 10.3389/fgene.2018.00360] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/21/2018] [Indexed: 12/26/2022] Open
Abstract
Pigs serve as an important agricultural resource and animal model in biomedical studies. Efficient and precise modification of pig genome by using recently developed gene editing tools has significantly broadened the application of pig models in various research areas. The three types of site-specific nucleases, namely, zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein, are the main gene editing tools that can efficiently introduce predetermined modifications, including knockouts and knockins, into the pig genome. These modifications can confer desired phenotypes to pigs to improve production traits, such as optimal meat production, enhanced feed digestibility, and disease resistance. Besides, given their genetic, anatomic, and physiologic similarities to humans, pigs can also be modified to model human diseases or to serve as an organ source for xenotransplantation to save human lives. To date, many genetically modified pig models with agricultural or biomedical values have been established by using gene editing tools. These pig models are expected to accelerate research progress in related fields and benefit humans.
Collapse
Affiliation(s)
- Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
43
|
White KA, Swier VJ, Cain JT, Kohlmeyer JL, Meyerholz DK, Tanas MR, Uthoff J, Hammond E, Li H, Rohret FA, Goeken A, Chan CH, Leidinger MR, Umesalma S, Wallace MR, Dodd RD, Panzer K, Tang AH, Darbro BW, Moutal A, Cai S, Li W, Bellampalli SS, Khanna R, Rogers CS, Sieren JC, Quelle DE, Weimer JM. A porcine model of neurofibromatosis type 1 that mimics the human disease. JCI Insight 2018; 3:120402. [PMID: 29925695 DOI: 10.1172/jci.insight.120402] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022] Open
Abstract
Loss of the NF1 tumor suppressor gene causes the autosomal dominant condition, neurofibromatosis type 1 (NF1). Children and adults with NF1 suffer from pathologies including benign and malignant tumors to cognitive deficits, seizures, growth abnormalities, and peripheral neuropathies. NF1 encodes neurofibromin, a Ras-GTPase activating protein, and NF1 mutations result in hyperactivated Ras signaling in patients. Existing NF1 mutant mice mimic individual aspects of NF1, but none comprehensively models the disease. We describe a potentially novel Yucatan miniswine model bearing a heterozygotic mutation in NF1 (exon 42 deletion) orthologous to a mutation found in NF1 patients. NF1+/ex42del miniswine phenocopy the wide range of manifestations seen in NF1 patients, including café au lait spots, neurofibromas, axillary freckling, and neurological defects in learning and memory. Molecular analyses verified reduced neurofibromin expression in swine NF1+/ex42del fibroblasts, as well as hyperactivation of Ras, as measured by increased expression of its downstream effectors, phosphorylated ERK1/2, SIAH, and the checkpoint regulators p53 and p21. Consistent with altered pain signaling in NF1, dysregulation of calcium and sodium channels was observed in dorsal root ganglia expressing mutant NF1. Thus, these NF1+/ex42del miniswine recapitulate the disease and provide a unique, much-needed tool to advance the study and treatment of NF1.
Collapse
Affiliation(s)
- Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | | | | | | | - Johanna Uthoff
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA
| | - Emily Hammond
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA
| | - Hua Li
- Department of Molecular Genetics and Microbiology and.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | | | | | - Chun-Hung Chan
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | | | | | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology and.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Rebecca D Dodd
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Karin Panzer
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Amy H Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA.,Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Song Cai
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Wennan Li
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | | | - Jessica C Sieren
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Dawn E Quelle
- Molecular Medicine Program.,Department of Pathology, and.,Department of Pharmacology and.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota, USA
| |
Collapse
|
44
|
van Leeuwen EM, Emri E, Merle BMJ, Colijn JM, Kersten E, Cougnard-Gregoire A, Dammeier S, Meester-Smoor M, Pool FM, de Jong EK, Delcourt C, Rodrigez-Bocanegra E, Biarnés M, Luthert PJ, Ueffing M, Klaver CCW, Nogoceke E, den Hollander AI, Lengyel I. A new perspective on lipid research in age-related macular degeneration. Prog Retin Eye Res 2018; 67:56-86. [PMID: 29729972 DOI: 10.1016/j.preteyeres.2018.04.006] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
There is an urgency to find new treatment strategies that could prevent or delay the onset or progression of AMD. Different classes of lipids and lipoproteins metabolism genes have been associated with AMD in a multiple ways, but despite the ever-increasing knowledge base, we still do not understand fully how circulating lipids or local lipid metabolism contribute to AMD. It is essential to clarify whether dietary lipids, systemic or local lipoprotein metabolismtrafficking of lipids in the retina should be targeted in the disease. In this article, we critically evaluate what has been reported in the literature and identify new directions needed to bring about a significant advance in our understanding of the role for lipids in AMD. This may help to develop potential new treatment strategies through targeting the lipid homeostasis.
Collapse
Affiliation(s)
- Elisabeth M van Leeuwen
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eszter Emri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Benedicte M J Merle
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Johanna M Colijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eveline Kersten
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Audrey Cougnard-Gregoire
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Sascha Dammeier
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Magda Meester-Smoor
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Eiko K de Jong
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Cécile Delcourt
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | | | | | | | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Everson Nogoceke
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Imre Lengyel
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
45
|
Burke AC, Telford DE, Sutherland BG, Edwards JY, Sawyez CG, Barrett PHR, Newton RS, Pickering JG, Huff MW. Bempedoic Acid Lowers Low-Density Lipoprotein Cholesterol and Attenuates Atherosclerosis in Low-Density Lipoprotein Receptor–Deficient (
LDLR
+/−
and
LDLR
−/−
) Yucatan Miniature Pigs. Arterioscler Thromb Vasc Biol 2018; 38:1178-1190. [DOI: 10.1161/atvbaha.117.310676] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/02/2018] [Indexed: 01/01/2023]
Abstract
Objective—
Bempedoic acid (BemA; ETC-1002) is a novel drug that targets hepatic ATP-citrate lyase to reduce cholesterol biosynthesis. In phase 2 studies, BemA lowers elevated low-density lipoprotein cholesterol (LDL-C) in hypercholesterolemic patients. In the present study, we tested the ability of BemA to decrease plasma cholesterol and LDL-C and attenuate atherosclerosis in a large animal model of familial hypercholesterolemia.
Approach and Results—
Gene targeting has been used to generate Yucatan miniature pigs heterozygous (
LDLR
+/−
) or homozygous (
LDLR
−/−
) for LDL receptor deficiency (ExeGen).
LDLR
+/−
and
LDLR
−/−
pigs were fed a high-fat, cholesterol-containing diet (34% kcal fat; 0.2% cholesterol) and orally administered placebo or BemA for 160 days. In
LDLR
+/−
pigs, compared with placebo, BemA decreased plasma cholesterol and LDL-C up to 40% and 61%, respectively. In
LDLR
−/−
pigs, in which plasma cholesterol and LDL-C were 5-fold higher than in
LDLR
+/−
pigs, BemA decreased plasma cholesterol and LDL-C up to 27% and 29%, respectively. Plasma levels of triglycerides and high-density lipoprotein cholesterol, fasting glucose and insulin, and liver lipids were unaffected by treatment in either genotype. In the aorta of
LDLR
+/−
pigs, BemA robustly attenuated en face raised lesion area (−58%) and left anterior descending coronary artery cross-sectional lesion area (−40%). In
LDLR
−/−
pigs, in which lesions were substantially more advanced, BemA decreased aortic lesion area (−47%) and left anterior descending coronary artery lesion area (−48%).
Conclusions—
In a large animal model of LDLR deficiency and atherosclerosis, long-term treatment with BemA reduces LDL-C and attenuates the development of aortic and coronary atherosclerosis in both
LDLR
+/−
and
LDLR
−/−
miniature pigs.
Collapse
Affiliation(s)
- Amy C. Burke
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Biochemistry (A.C.B., J.G.P., M.W.H.)
| | - Dawn E. Telford
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Medicine (D.E.T., J.Y.E., C.G.S., J.G.P., M.W.H.), The University of Western Ontario, London, Canada
| | - Brian G. Sutherland
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
| | - Jane Y. Edwards
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Medicine (D.E.T., J.Y.E., C.G.S., J.G.P., M.W.H.), The University of Western Ontario, London, Canada
| | - Cynthia G. Sawyez
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Medicine (D.E.T., J.Y.E., C.G.S., J.G.P., M.W.H.), The University of Western Ontario, London, Canada
| | - P. Hugh R. Barrett
- School of Biomedical Sciences, University of Western Australia, Perth (P.H.R.B.)
| | | | - J. Geoffrey Pickering
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Biochemistry (A.C.B., J.G.P., M.W.H.)
- Department of Medicine (D.E.T., J.Y.E., C.G.S., J.G.P., M.W.H.), The University of Western Ontario, London, Canada
| | - Murray W. Huff
- From the Robarts Research Institute (A.C.B., D.E.T., B.G.S., J.Y.E., C.G.S., J.G.P., M.W.H.)
- Department of Biochemistry (A.C.B., J.G.P., M.W.H.)
- Department of Medicine (D.E.T., J.Y.E., C.G.S., J.G.P., M.W.H.), The University of Western Ontario, London, Canada
| |
Collapse
|
46
|
Huang L, Hua Z, Xiao H, Cheng Y, Xu K, Gao Q, Xia Y, Liu Y, Zhang X, Zheng X, Mu Y, Li K. CRISPR/Cas9-mediated ApoE-/- and LDLR-/- double gene knockout in pigs elevates serum LDL-C and TC levels. Oncotarget 2018; 8:37751-37760. [PMID: 28465483 PMCID: PMC5514946 DOI: 10.18632/oncotarget.17154] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
The traditional method to establish a cardiovascular disease model induced by high fat and high cholesterol diets is time consuming and laborious and may not be appropriate in all circumstances. A suitable pig model to study metabolic disorders and subsequent atherosclerosis is not currently available. For this purpose, we applied the CRISPR/Cas9 system to Bama minipigs, targeting apolipoprotein E (ApoE) and low density lipoprotein receptor (LDLR) gene simultaneously. Six biallelic knockout pigs of these two genes were obtained successfully in a single step. No off-target incidents or mosaic mutations were detected by an unbiased analysis. Serum biochemical analyses of gene-modified piglets showed that the levels of low density lipoprotein choleserol (LDL-C), total cholesterol (TC) and apolipoprotein B (APOB) were elevated significantly. This model should prove valuable for the study of human cardiovascular disease and related translational research.
Collapse
Affiliation(s)
- Lei Huang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,Animal Functional Genomics Group, Agricultural Genomes Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Zaidong Hua
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Hongwei Xiao
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Ying Cheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qian Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ying Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xinming Zheng
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Yulian Mu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
47
|
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: 2.3] [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
|
48
|
Kleinert M, Clemmensen C, Hofmann SM, Moore MC, Renner S, Woods SC, Huypens P, Beckers J, de Angelis MH, Schürmann A, Bakhti M, Klingenspor M, Heiman M, Cherrington AD, Ristow M, Lickert H, Wolf E, Havel PJ, Müller TD, Tschöp MH. Animal models of obesity and diabetes mellitus. Nat Rev Endocrinol 2018; 14:140-162. [PMID: 29348476 DOI: 10.1038/nrendo.2017.161] [Citation(s) in RCA: 536] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
More than one-third of the worldwide population is overweight or obese and therefore at risk of developing type 2 diabetes mellitus. In order to mitigate this pandemic, safer and more potent therapeutics are urgently required. This necessitates the continued use of animal models to discover, validate and optimize novel therapeutics for their safe use in humans. In order to improve the transition from bench to bedside, researchers must not only carefully select the appropriate model but also draw the right conclusions. In this Review, we consolidate the key information on the currently available animal models of obesity and diabetes and highlight the advantages, limitations and important caveats of each of these models.
Collapse
Affiliation(s)
- Maximilian Kleinert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Susanna M Hofmann
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Ziemssenstr. 1, D-80336 Munich, Germany
| | - Mary C Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| | - Simone Renner
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilan University München, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Stephen C Woods
- University of Cincinnati College of Medicine, Department of Psychiatry and Behavioral Neuroscience, Metabolic Diseases Institute, 2170 East Galbraith Road, Cincinnati, Ohio 45237, USA
| | - Peter Huypens
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Johannes Beckers
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, D-85354 Freising, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, D-85354 Freising, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Mostafa Bakhti
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technische Universität München, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
- Else Kröner-Fresenius Center for Nutritional Medicine, Technische Universität München, D-85354 Freising, Germany
- Institute for Food & Health, Technische Universität München, D-85354 Freising, Germany
| | - Mark Heiman
- MicroBiome Therapeutics, 1316 Jefferson Ave, New Orleans, Louisiana 70115, USA
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, CH-8603 Zurich-Schwerzenbach, Switzerland
| | - Heiko Lickert
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilan University München, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, 3135 Meyer Hall, University of California, Davis, California 95616-5270, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| |
Collapse
|
49
|
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: 1.8] [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
|
50
|
Jakobsen JE, Johansen MG, Schmidt M, Liu Y, Li R, Callesen H, Melnikova M, Habekost M, Matrone C, Bouter Y, Bayer TA, Nielsen AL, Duthie M, Fraser PE, Holm IE, Jørgensen AL. Expression of the Alzheimer's Disease Mutations AβPP695sw and PSEN1M146I in Double-Transgenic Göttingen Minipigs. J Alzheimers Dis 2018; 53:1617-30. [PMID: 27540966 DOI: 10.3233/jad-160408] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mutations in the amyloid-β protein precursor gene (AβPP), the presenilin 1 gene (PSEN1) or the presenilin 2 gene (PSEN2) that increase production of the AβPP-derived peptide Aβ42 cause early-onset Alzheimer's disease. Rodent models of the disease show that further increase in Aβ42 production and earlier brain pathology can be obtained by coexpressing AβPP and PSEN1 mutations. To generate such elevated Aβ42 level in a large animal model, we produced Göttingen minipigs carrying in their genome one copy of a human PSEN1 cDNA with the Met146Ile (PSEN1M146I) mutation and three copies of a human AβPP695 cDNA with the Lys670Asn/Met671Leu (AβPPsw) double-mutation. Both transgenes were expressed in fibroblasts and in the brain, and their respective proteins were processed normally. Immunohistochemical staining with Aβ42-specific antibodies detected intraneuronal accumulation of Aβ42 in brains from a 10- and an 18-month-old pig. Such accumulation may represent an early event in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jannik E Jakobsen
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | | | - Mette Schmidt
- Department of Large Animal Sciences/Reproduction, University of Copenhagen, Frederiksberg C, Denmark
| | - Ying Liu
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | - Rong Li
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | - Henrik Callesen
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | | | - Mette Habekost
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | - Carmela Matrone
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | | | - Monika Duthie
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Ida E Holm
- Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark.,Department of Pathology, Randers Hospital, Randers, Denmark
| | | |
Collapse
|