1
|
Cheng SH, Chu W, Chou WH, Chu WC, Kang YN. Cardiovascular Safety of Romosozumab Compared to Commonly Used Anti-osteoporosis Medications in Postmenopausal Osteoporosis: A Systematic Review and Network Meta-analysis of Randomized Controlled Trials. Drug Saf 2025; 48:7-23. [PMID: 39227560 DOI: 10.1007/s40264-024-01475-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION The aim of this study was to investigate the cardiovascular safety of romosozumab in postmenopausal women with osteoporosis. Romosozumab, a monoclonal antibody targeting sclerostin, has been shown to increase bone mineral density and reduce the risk of osteoporotic fractures. However, in previous studies, romosozumab therapy was identified as a potential risk factor for cardiovascular events, particularly in patients with predisposing cardiovascular disease. METHODS A systematic literature search was performed in the Cochrane Library, Embase, PubMed, and Web of Science databases to identify randomized controlled trials (RCTs) comparing the safety and efficacy of romosozumab versus alendronate, teriparatide, denosumab, or placebo in postmenopausal women with osteoporosis. Contrast-based network meta-analysis was performed using a random-effects model. The pooled estimates are presented as risk ratios with 95% confidence intervals. RESULTS Of the 5282 articles retrieved, 25 RCTs were included in this review (n = 24,942), and 18 randomized controlled trials (n = 16,777) were included in the network meta-analysis. The results indicated no significant differences in cardiovascular mortality rate between romosozumab and placebo. Regarding the risk of major cardiovascular events, no significant differences were found in the direct evidence or the network meta-analysis with placebo as the reference. CONCLUSION Romosozumab might be a safe option for treating postmenopausal women with osteoporosis. The cardiovascular concerns associated with this treatment seem less significant than previously suggested, although additional real-world data are required to confirm this conclusion.
Collapse
Affiliation(s)
- Shih-Hao Cheng
- Department of Biomedical Engineering, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
- Department of Orthopedics, Wan Fang Hospital, Medical University Hospital, Taipei, Taiwan
| | - William Chu
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Wen-Hsiang Chou
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Woei-Chyn Chu
- Department of Biomedical Engineering, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.
| | - Yi-No Kang
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
- Evidence-Based Medicine Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Institute of Health Policy and Management, College of Public Health, National Taiwan University, Taipei, Taiwan.
- Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Odogwu NM, Jang JS, Albertson S, Hagen C, Rasmussen B, Saji O, Nelson TJ. Optimizing RNA extraction methods for high-throughput transcriptome sequencing of formalin-fixed paraffin-embedded cardiac tissue specimens. PLoS One 2024; 19:e0315098. [PMID: 39724161 DOI: 10.1371/journal.pone.0315098] [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: 10/01/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Archived FFPE cardiac tissue specimens are valuable for molecular studies aimed at identifying biomarkers linked to mortality in cardiovascular disease. Establishing a reliable and reproducible RNA extraction method is critical for generating high-quality transcriptome sequences for molecular assays. Here, the efficiency of four RNA extraction methods: Qiagen AllPrep DNA/RNA method (Method QP); Qiagen AllPrep DNA/RNA method with protocol modification on the ethanol wash step after deparaffinization (Method QE); CELLDATA RNA extraction (Method BP) and CELLDATA RNA extraction with protocol modifications on the lysis step (Method BL) was compared on 23 matching FFPE cardiac tissue specimens (n = 92).In comparing RNA quality metrics across FFPE RNA extract, nucleic acids extracted deploying Method QE and QP produced the highest RNA yield. However, Method QE outperformed Method QP as more extract from Method QE had DV 200 values above 30%. Both method BL and BP produced similar range of RNA purity and yield but more extract from Method BL had DV 200 values above 30% compared to Method BP. When accessing distribution value, Method BL outperformed Methods BP, QE, and QP as more extracts from Method BL had DV 200 values above 30% compared to other methods (PDV200<0.001; Kruskal-Wallis). Method QE outperformed other methods in terms of RNA yield. RNA extracts from Method QE, characterized by high RNA yield, achieved sequencing results comparable to those from Method BL, characterized by high DV200 values. Our findings reveal that optimizing protocols can yield higher-quality RNA, facilitating the exploration of more disease conditions with high-resolution transcriptome profiling.
Collapse
Affiliation(s)
- Nkechi Martina Odogwu
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Jin Sung Jang
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Sabrina Albertson
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Clinton Hagen
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Boyd Rasmussen
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Oommen Saji
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| | - Timothy J Nelson
- Program for Hypoplastic Left Heart Syndrome, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
- Division of Cardiovascular Medicine, General Internal Medicine, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
- Center for Regenerative Medicine, Mayo Clinic Rochester, Rochester, Minnesota, United States of America
| |
Collapse
|
3
|
Tepavčević S, Zec M, Stojiljković M, Bošković M, Ćulafić T, Stanković A, Romić S, Živković M, Korićanac G. Unlocking the Cardiovascular Benefits of Walnuts: Insights on Molecular Mechanism From Animal Studies. Nutr Rev 2024:nuae173. [PMID: 39565929 DOI: 10.1093/nutrit/nuae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
Abstract
The health-promoting benefits of walnut consumption are substantially ascribed to its fatty acid (FA) profile, which is rich in polyunsaturated FA with an exceptionally high n-3 to n-6 ratio. There are also phytonutrients in walnuts that are linked to health welfare. This review article integrates all studies on the effects of walnuts on the cardiovascular system performed on experimental animals, and thus is a source of data on the mechanisms underlying the observed effects. These studies, which are very diverse in experimental design, indicate that a diet enriched with walnuts or treating animals with walnut extract or chemical constituents of walnuts, has many favorable effects on heart and vascular system function. The cardiovascular effect of walnuts depends on the metabolic status of the organism. Among the cardiovascular effects of walnuts is that they improve the FA profile in the circulation and heart in favor of n-3 polyunsaturated FAs. In addition, a favorable effect on triglyceride and cholesterol status, which reduces cardiovascular disease risk, is observed. Intake of walnuts promotes FA catabolism and has anti-inflammatory, antioxidant, and antiarrhythmic effects. Walnuts also have a beneficial effect on vascular tone, accompanied by a decrease in blood pressure and reduced risk for atherosclerosis. In conclusion, studies on experimental animals encourage the consumption of walnuts as a simple, convenient approach to improve cardiovascular health.
Collapse
Affiliation(s)
- Snežana Tepavčević
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Manja Zec
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Mojca Stojiljković
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Maja Bošković
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Tijana Ćulafić
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Aleksandra Stanković
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Snježana Romić
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Maja Živković
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Goran Korićanac
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| |
Collapse
|
4
|
Sleiman Y, Reisqs JB, Boutjdir M. Differentiation of Sinoatrial-like Cardiomyocytes as a Biological Pacemaker Model. Int J Mol Sci 2024; 25:9155. [PMID: 39273104 PMCID: PMC11394733 DOI: 10.3390/ijms25179155] [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: 07/20/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are widely used for disease modeling and pharmacological screening. However, their application has mainly focused on inherited cardiopathies affecting ventricular cardiomyocytes, leading to extensive knowledge on generating ventricular-like hiPSC-CMs. Electronic pacemakers, despite their utility, have significant disadvantages, including lack of hormonal responsiveness, infection risk, limited battery life, and inability to adapt to changes in heart size. Therefore, developing an in vitro multiscale model of the human sinoatrial node (SAN) pacemaker using hiPSC-CM and SAN-like cardiomyocyte differentiation protocols is essential. This would enhance the understanding of SAN-related pathologies and support targeted therapies. Generating SAN-like cardiomyocytes offers the potential for biological pacemakers and specialized conduction tissues, promising significant benefits for patients with conduction system defects. This review focuses on arrythmias related to pacemaker dysfunction, examining protocols' advantages and drawbacks for generating SAN-like cardiomyocytes from hESCs/hiPSCs, and discussing therapeutic approaches involving their engraftment in animal models.
Collapse
Affiliation(s)
- Yvonne Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
| | - Jean-Baptiste Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
5
|
Ali S, Zulfiqar M, Summer M, Arshad M, Noor S, Nazakat L, Javed A. Zebrafish as an innovative model for exploring cardiovascular disease induction mechanisms and novel therapeutic interventions: a molecular insight. Mol Biol Rep 2024; 51:904. [PMID: 39133413 DOI: 10.1007/s11033-024-09814-w] [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: 06/03/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Cardiovascular disease (CVD) is a common cardiac disorder that leads to heart attacks, strokes, and heart failure. It is primarily characterized by conditions that impact the heart and blood arteries, including peripheral artery disease, arrhythmias, atherosclerosis, myocardial ischemia, congenital heart abnormalities, heart failure, rheumatic heart disease, hypertension, and cardiomyopathies. These conditions are mainly effect the heart and blood vessels, causing blockages or weakened pumping, due to severe hereditary and environmental factors. The frequency of CVD is rising significantly as life expectancy increases. Despite this, no effective treatment or management for its symptoms has been found. One of the most difficult obstacles to overcome, is finding a suitable animal model for drug screening and drug development. Although rodents, mice, swine, and mammals serve as the basis for most animal models of cardiovascular disease, no model accurately captures the epidemiology of the condition. Zebrafish (Danio rerio) have drawn the interest of the international scientific community due to certain shortcomings of the previously discussed animal models because they are smaller, less costly, and have an incredibly high rate of reproduction. This review article emphasizes the significance of using zebrafish as an animal model to investigate the possible facets of cardiovascular disease. Moreover, the ultimate purpose of this review article is to establish the advantages of employing zebrafish over other animal models and to investigate the boundaries of using zebrafish to study human disease. Furthermore, the mechanisms of cardiovascular diseases induction in zebrafish were covered to improve understanding for readers. Finally, the analysis of cardiotoxicity using Zebra fish model, is also explained. In order to stop the health index from deteriorating, the current study also covers some innovative, effective, and relatively safer treatments for treatment and management of cardiotoxicity.
Collapse
Affiliation(s)
- Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Maryam Zulfiqar
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Mahnoor Arshad
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Laiba Nazakat
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Abdullah Javed
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
6
|
Sun F, Chen X, Zhang S, Jiang H, Chen T, Xing T, Li X, Sultan R, Wang Z, Jia J. Cross-species signaling pathways analysis inspire animal model selections for drug screening and target prediction in vascular aging diseases. Evol Appl 2024; 17:e13708. [PMID: 38863828 PMCID: PMC11164676 DOI: 10.1111/eva.13708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/03/2024] [Indexed: 06/13/2024] Open
Abstract
Age is a significant contributing factor to the occurrence and progression of cardiovascular disease (CVD). Pharmacological treatment can effectively alleviate CVD symptoms caused by aging. However, 90% of the drugs have failed in clinics because of the loss of drug effects or the occurrence of the side effects. One of the reasons is the disparity between animal models used and the actual physiological levels in humans. Therefore, we integrated multiple datasets from single-cell and bulk-seq RNA-sequencing data in rats, monkeys, and humans to identify genes and pathways with consistent/differential expression patterns across these three species. An approach called "Cross-species signaling pathway analysis" was developed to select suitable animal models for drug screening. The effectiveness of this method was validated through the analysis of the pharmacological predictions of four known anti-vascular aging drugs used in animal/clinical experiments. The effectiveness of drugs was consistently observed between the models and clinics when they targeted pathways with the same trend in our analysis. However, drugs might have exhibited adverse effects if they targeted pathways with opposite trends between the models and the clinics. Additionally, through our approach, we discovered four targets for anti-vascular aging drugs, which were consistent with their pharmaceutical effects in literatures, showing the value of this approach. In the end, software was established to facilitate the use of "Cross-species signaling pathway analysis." In sum, our study suggests utilizing bioinformatics analysis based on disease characteristics can help in choosing more appropriate animal models.
Collapse
Affiliation(s)
- Fei Sun
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Xingxing Chen
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Shuqing Zhang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Haihong Jiang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Tianhong Chen
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Tongying Xing
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Xueyi Li
- Sino‐Swiss Institute of Advanced Technology, School of Micro‐ElectronicsShanghai UniversityShanghaiChina
| | - Rabia Sultan
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Zhimin Wang
- Shanghai‐MOST Key Laboratory of Health and Disease GenomicsShanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghaiChina
| | - Jia Jia
- School of Life SciencesShanghai UniversityShanghaiChina
- Sino‐Swiss Institute of Advanced Technology, School of Micro‐ElectronicsShanghai UniversityShanghaiChina
| |
Collapse
|
7
|
Gola C, Fingerhood S, Parry NM, Diaz-Delgado J. Dissecting aortitis in a goat associated with Pasteurella multocida and Staphylococcus spp infection. J Comp Pathol 2024; 211:17-20. [PMID: 38759507 DOI: 10.1016/j.jcpa.2024.04.002] [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: 02/03/2024] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 05/19/2024]
Abstract
Reports of primary cardiovascular disease in goats are rare and most commonly include ventricular septal defect, valvular endocarditis, traumatic pericarditis, ionophore poisoning and nutritional cardiomyopathies. We now report the pathological findings in a 67 kg, 6-year-old, adult female Boer goat that presented with neurological signs (ie, head pressing, unsteadiness and paddling) and hyperthermia 2 days prior to death. Lack of therapeutic response to meloxicam and penicillin‒streptomycin and poor prognosis led to euthanasia of the animal. At necropsy, the main findings included severe aortic dissection with luminal thrombosis and stenosis, and pulmonary congestion and oedema. Histological examination of the aorta revealed severe chronic granulomatous and fibrosing dissecting aortitis with mineralization. Bacterial culture of the affected aortic segment resulted in isolation of a profuse growth of Pasteurella multocida and a moderate growth of Staphylococcus spp. Histopathological findings in the central nervous system were consistent with neurolisteriosis.
Collapse
Affiliation(s)
- Cecilia Gola
- Veterinary Pathology Centre, University of Surrey, Francis Crick Road, Guildford, GU27AQ, Surrey, UK
| | - Sai Fingerhood
- Veterinary Pathology Centre, University of Surrey, Francis Crick Road, Guildford, GU27AQ, Surrey, UK.
| | - Nicola M Parry
- CBSET Inc., 500 Shire Way, Lexington, Massachussetts, 02421, USA
| | - Josué Diaz-Delgado
- Texas A&M Veterinary Medical Diagnostic Laboratory, 483 Agronomy Road, College Station, Texas, 77843, USA
| |
Collapse
|
8
|
Calvo-Sánchez N, Rodríguez-Largo A, Puzol L, de Miguel R, Pérez E, Gómez Á, Micheloud JF, Luján L. Corrugated intimal surface of the ovine aorta: when physiology resembles pathology. J Comp Pathol 2024; 211:8-11. [PMID: 38636282 DOI: 10.1016/j.jcpa.2024.03.207] [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: 11/17/2023] [Revised: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
The aortic lumen in healthy animals is characterized by a smooth, whitish surface, but sheep have macroscopic corrugation of the intimal surface in the thoracic aorta (TA). Our aim was to determine if this finding was pathological or physiological. Thirteen sheep aortas were included in this work together with aortas from cattle (n = 3), a goat (n = 1), horses (n = 4), dogs (n = 2), rabbits (n = 2) and a pig (n = 1). A corrugated intimal surface in the TA was seen in all the sheep and the goat but was less evident in the cattle. Histologically, in sheep the TA intimal surface was seen to have multifocal bulging areas that protruded into the lumen. The outer half of the tunica media had numerous, randomly distributed muscle islands that disrupted the arrangement of the elastic lamella, displacing them towards the lumen. We conclude that the intimal corrugation of the TA in sheep is physiological and must not be misinterpreted as pathological.
Collapse
Affiliation(s)
- Natalia Calvo-Sánchez
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain; AgriFood Institute of Aragon, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Ana Rodríguez-Largo
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Leonor Puzol
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Ricardo de Miguel
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Estela Pérez
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain; AgriFood Institute of Aragon, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Álex Gómez
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain; AgriFood Institute of Aragon, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain
| | - Juan F Micheloud
- Instituto Nacional de Tecnología Agropecuaria, RN 68, km 72, Cerrillos, Salta 4403, Argentina; Universidad Católica de Salta, Campus Castañares, A4400 Salta, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Goday Cruz 2290, Buenos Aires, Argentina
| | - Lluís Luján
- Department of Animal Pathology, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain; AgriFood Institute of Aragon, Veterinary Faculty, Universidad de Zaragoza, Miguel Servet Street, 177, 50013 Zaragoza, Spain.
| |
Collapse
|
9
|
van Rhijn-Brouwer FCCC, Wever KE, Kiffen R, van Rhijn JR, Gremmels H, Fledderus JO, Vernooij RWM, Verhaar MC. Systematic review and meta-analysis of the effect of bone marrow-derived cell therapies on hind limb perfusion. Dis Model Mech 2024; 17:dmm050632. [PMID: 38616715 PMCID: PMC11139036 DOI: 10.1242/dmm.050632] [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: 11/27/2023] [Accepted: 04/03/2024] [Indexed: 04/16/2024] Open
Abstract
Preclinical and clinical studies on the administration of bone marrow-derived cells to restore perfusion show conflicting results. We conducted a systematic review and meta-analysis on preclinical studies to assess the efficacy of bone marrow-derived cells in the hind limb ischemia model and identify possible determinants of therapeutic efficacy. In vivo animal studies were identified using a systematic search in PubMed and EMBASE on 10 January 2022. 85 studies were included for systematic review and meta-analysis. Study characteristics and outcome data on relative perfusion were extracted. The pooled mean difference was estimated using a random effects model. Risk of bias was assessed for all included studies. We found a significant increase in perfusion in the affected limb after administration of bone marrow-derived cells compared to that in the control groups. However, there was a high heterogeneity between studies, which could not be explained. There was a high degree of incomplete reporting across studies. We therefore conclude that the current quality of preclinical research is insufficient (low certainty level as per GRADE assessment) to identify specific factors that might improve human clinical trials.
Collapse
Affiliation(s)
| | - Kimberley Elaine Wever
- Department of Anaesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Romy Kiffen
- Department of Anaesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jon-Ruben van Rhijn
- Institute of Life Sciences and Chemistry, HU University of Applied Sciences Utrecht, 3584 CS Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Joost Ougust Fledderus
- Department of Nephrology and Hypertension, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Robin Wilhelmus Maria Vernooij
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Marianne Christina Verhaar
- Department of Nephrology and Hypertension, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
10
|
Stone CR, Harris DD, Broadwin M, Kanuparthy M, Sabe SA, Xu C, Feng J, Abid MR, Sellke FW. Crafting a Rigorous, Clinically Relevant Large Animal Model of Chronic Myocardial Ischemia: What Have We Learned in 20 Years? Methods Protoc 2024; 7:17. [PMID: 38392691 PMCID: PMC10891802 DOI: 10.3390/mps7010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/10/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
The past several decades have borne witness to several breakthroughs and paradigm shifts within the field of cardiovascular medicine, but one component that has remained constant throughout this time is the need for accurate animal models for the refinement and elaboration of the hypotheses and therapies crucial to our capacity to combat human disease. Numerous sophisticated and high-throughput molecular strategies have emerged, including rational drug design and the multi-omics approaches that allow extensive characterization of the host response to disease states and their prospective resolutions, but these technologies all require grounding within a faithful representation of their clinical context. Over this period, our lab has exhaustively tested, progressively refined, and extensively contributed to cardiovascular discovery on the basis of one such faithful representation. It is the purpose of this paper to review our porcine model of chronic myocardial ischemia using ameroid constriction and the subsequent myriad of physiological and molecular-biological insights it has allowed our lab to attain and describe. We hope that, by depicting our methods and the insight they have yielded clearly and completely-drawing for this purpose on comprehensive videographic illustration-other research teams will be empowered to carry our work forward, drawing on our experience to refine their own investigations into the pathogenesis and eradication of cardiovascular disease.
Collapse
Affiliation(s)
- Christopher R. Stone
- Department of Cardiothoracic Surgery, The Warren Alpert School of Medicine at Brown University, Providence, RI 02903, USA; (D.D.H.); (M.B.); (M.K.); (S.A.S.); (C.X.); (J.F.); (M.R.A.); (F.W.S.)
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Summers KM. Genetic models of fibrillinopathies. Genetics 2024; 226:iyad189. [PMID: 37972149 PMCID: PMC11021029 DOI: 10.1093/genetics/iyad189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
The fibrillinopathies represent a group of diseases in which the 10-12 nm extracellular microfibrils are disrupted by genetic variants in one of the genes encoding fibrillin molecules, large glycoproteins of the extracellular matrix. The best-known fibrillinopathy is Marfan syndrome, an autosomal dominant condition affecting the cardiovascular, ocular, skeletal, and other systems, with a prevalence of around 1 in 3,000 across all ethnic groups. It is caused by variants of the FBN1 gene, encoding fibrillin-1, which interacts with elastin to provide strength and elasticity to connective tissues. A number of mouse models have been created in an attempt to replicate the human phenotype, although all have limitations. There are also natural bovine models and engineered models in pig and rabbit. Variants in FBN2 encoding fibrillin-2 cause congenital contractural arachnodactyly and mouse models for this condition have also been produced. In most animals, including birds, reptiles, and amphibians, there is a third fibrillin, fibrillin-3 (FBN3 gene) for which the creation of models has been difficult as the gene is degenerate and nonfunctional in mice and rats. Other eukaryotes such as the nematode C. elegans and zebrafish D. rerio have a gene with some homology to fibrillins and models have been used to discover more about the function of this family of proteins. This review looks at the phenotype, inheritance, and relevance of the various animal models for the different fibrillinopathies.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba QLD 4102, Australia
| |
Collapse
|
12
|
Dittfeld C, Winkelkotte M, Scheer A, Voigt E, Schmieder F, Behrens S, Jannasch A, Matschke K, Sonntag F, Tugtekin SM. Challenges of aortic valve tissue culture - maintenance of viability and extracellular matrix in the pulsatile dynamic microphysiological system. J Biol Eng 2023; 17:60. [PMID: 37770970 PMCID: PMC10538250 DOI: 10.1186/s13036-023-00377-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/14/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) causes an increasing health burden in the 21st century due to aging population. The complex pathophysiology remains to be understood to develop novel prevention and treatment strategies. Microphysiological systems (MPSs), also known as organ-on-chip or lab-on-a-chip systems, proved promising in bridging in vitro and in vivo approaches by applying integer AV tissue and modelling biomechanical microenvironment. This study introduces a novel MPS comprising different micropumps in conjunction with a tissue-incubation-chamber (TIC) for long-term porcine and human AV incubation (pAV, hAV). RESULTS Tissue cultures in two different MPS setups were compared and validated by a bimodal viability analysis and extracellular matrix transformation assessment. The MPS-TIC conjunction proved applicable for incubation periods of 14-26 days. An increased metabolic rate was detected for pulsatile dynamic MPS culture compared to static condition indicated by increased LDH intensity. ECM changes such as an increase of collagen fibre content in line with tissue contraction and mass reduction, also observed in early CAVD, were detected in MPS-TIC culture, as well as an increase of collagen fibre content. Glycosaminoglycans remained stable, no significant alterations of α-SMA or CD31 epitopes and no accumulation of calciumhydroxyapatite were observed after 14 days of incubation. CONCLUSIONS The presented ex vivo MPS allows long-term AV tissue incubation and will be adopted for future investigation of CAVD pathophysiology, also implementing human tissues. The bimodal viability assessment and ECM analyses approve reliability of ex vivo CAVD investigation and comparability of parallel tissue segments with different treatment strategies regarding the AV (patho)physiology.
Collapse
Affiliation(s)
- Claudia Dittfeld
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany.
| | - Maximilian Winkelkotte
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Anna Scheer
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Emmely Voigt
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Florian Schmieder
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Stephan Behrens
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Anett Jannasch
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Klaus Matschke
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Frank Sonntag
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Sems-Malte Tugtekin
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| |
Collapse
|
13
|
Cassaday HJ, Cavenagh L, Aluthgamage H, Crooks A, Bonardi C, Stevenson CW, Waite L, Muir C. Attitudes to the use of animals in biomedical research: Effects of stigma and selected research project summaries. PLoS One 2023; 18:e0290232. [PMID: 37594971 PMCID: PMC10437917 DOI: 10.1371/journal.pone.0290232] [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: 03/22/2023] [Accepted: 08/06/2023] [Indexed: 08/20/2023] Open
Abstract
Three groups of participants (largely recruited from the UK) completed a survey to examine attitudes to the use of animals in biomedical research, after reading the lay (N = 182) or technical (N = 201) summary of a research project, or no summary (N = 215). They then completed a survey comprising the animal attitude (AAS), animal purpose (APQ), belief in animal mind (BAM) and empathy quotient (EQ) scales. The APQ was adapted to assess attitudes towards the use of animals for research into disorders selected to be perceived as controllable and so 'blameworthy' and potentially stigmatised (addiction and obesity) and 'psychological' (schizophrenia and addiction) versus 'physical' (cardiovascular disease and obesity), across selected species (rats, mice, fish pigs and monkeys). Thus, the APQ was used to examine how the effects of perceived controllability and the nature of the disorder affected attitudes to animal use, in different species and in the three summary groups. As expected, attitudes to animal use as measured by the AAS and the APQ (total) correlated positively with BAM and EQ scores, consistent with the assumption that the scales all measured pro-welfare attitudes. Participants in the two research summary groups did not differentiate the use of rats, mice and fish (or fish and pigs in the technical summary group), whereas all species were differentiated in the no summary group. Participants given the lay summary were as concerned about the use of animals for schizophrenia as for addiction research. APQ ratings otherwise indicated more concern for animals used for addiction research (and for obesity compared to cardiovascular disease in all summary groups). Therefore, the information provided by a research project summary influenced attitudes to use of animals in biomedical research. However, there was no overall increase in agreement with animal use in either of the summary groups.
Collapse
Affiliation(s)
- Helen J. Cassaday
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Lucy Cavenagh
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Hiruni Aluthgamage
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Aoife Crooks
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte Bonardi
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Carl W. Stevenson
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Lauren Waite
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte Muir
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
14
|
Bayne EF, Rossler KJ, Gregorich ZR, Aballo TJ, Roberts DS, Chapman EA, Guo W, Palecek SP, Ralphe JC, Kamp TJ, Ge Y. Top-down proteomics of myosin light chain isoforms define chamber-specific expression in the human heart. J Mol Cell Cardiol 2023; 181:89-97. [PMID: 37327991 PMCID: PMC10528938 DOI: 10.1016/j.yjmcc.2023.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/27/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Myosin functions as the "molecular motor" of the sarcomere and generates the contractile force necessary for cardiac muscle contraction. Myosin light chains 1 and 2 (MLC-1 and -2) play important functional roles in regulating the structure of the hexameric myosin molecule. Each of these light chains has an 'atrial' and 'ventricular' isoform, so called because they are believed to exhibit chamber-restricted expression in the heart. However, recently the chamber-specific expression of MLC isoforms in the human heart has been questioned. Herein, we analyzed the expression of MLC-1 and -2 atrial and ventricular isoforms in each of the four cardiac chambers in adult non-failing donor hearts using top-down mass spectrometry (MS)-based proteomics. Strikingly, we detected an isoform thought to be ventricular, MLC-2v (gene: MYL2), in the atria and confirmed the protein sequence using tandem MS (MS/MS). For the first time, a putative deamidation post-translation modification (PTM) located on MLC-2v in atrial tissue was localized to amino acid N13. MLC-1v (MYL3) and MLC-2a (MYL7) were the only MLC isoforms exhibiting chamber-restricted expression patterns across all donor hearts. Importantly, our results unambiguously show that MLC-1v, not MLC-2v, is ventricle-specific in adult human hearts. Moreover, we found elevated MLC-2 phosphorylation in male hearts compared to female hearts across each cardiac chamber. Overall, top-down proteomics allowed an unbiased analysis of MLC isoform expression throughout the human heart, uncovering previously unexpected isoform expression patterns and PTMs.
Collapse
Affiliation(s)
- Elizabeth F Bayne
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kalina J Rossler
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachery R Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Timothy J Aballo
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Emily A Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
15
|
Alkhatib B, Salimi S, Jabari M, Padmanabhan V, Vyas AK. Impact of Adverse Gestational Milieu on Maternal Cardiovascular Health. Endocrinology 2023; 164:bqad060. [PMID: 37042476 PMCID: PMC10164662 DOI: 10.1210/endocr/bqad060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/13/2023]
Abstract
Cardiovascular disease affects 1% to 4% of the nearly 4 million pregnancies in the United States each year and is the primary cause of pregnancy-related mortality. Adverse pregnancy outcomes are associated with cardiovascular complications during pregnancy persisting into the postpartum period. Recently, investigations have identified an altered sex hormone milieu, such as in the case of hyperandrogenism, as a causative factor in the development of gestational cardiovascular dysfunction. The mechanisms involved in the development of cardiovascular disease in postpartum women are largely unknown. Animal studies have attempted to recapitulate adverse pregnancy outcomes to investigate causal relationships and molecular underpinnings of adverse gestational cardiac events and progression to the development of cardiovascular disease postpartum. This review will focus on summarizing clinical and animal studies detailing the impact of adverse pregnancy outcomes, including preeclampsia, gestational diabetes mellitus, and maternal obesity, on gestational cardiometabolic dysfunction and postpartum cardiovascular disease. Specifically, we will highlight the adverse impact of gestational hyperandrogenism and its potential to serve as a biomarker for maternal gestational and postpartum cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St. Louis, MO 63110, USA
| | - Shadi Salimi
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Mary Jabari
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | | | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St. Louis, MO 63110, USA
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
16
|
Roths M, Freestone AD, Rudolph TE, Michael A, Baumgard LH, Selsby JT. Environment-induced heat stress causes structural and biochemical changes in the heart. J Therm Biol 2023; 113:103492. [PMID: 37055111 DOI: 10.1016/j.jtherbio.2023.103492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/07/2023]
Abstract
Prolonged exposure to heat can lead to environment-induced heat stress (EIHS), which may jeopardize human health, but the extent to which EIHS affects cardiac architecture and myocardial cell health are unknown. We hypothesized EIHS would alter cardiac structure and cause cellular dysfunction. To test this hypothesis, 3-mo old female pigs were exposed to thermoneutral (TN; 20.6 ± 0.2 °C; n = 8) or EIHS (37.4 ± 0.2 °C; n = 8) conditions for 24 h, hearts were removed and dimensions measured, and portions of the left ventricle (LV) and right ventricle (RV) were collected. Environment-induced heat stress increased rectal temperature 1.3 °C (P < 0.01), skin temperature 11 °C (P < 0.01) and respiratory rate 72 breaths per minute (P < 0.01). Heart weight and length (apex to base) were decreased by 7.6% (P = 0.04) and 8.5% (P = 0.01), respectively, by EIHS, but heart width was similar between groups. Left ventricle wall thickness was increased (22%; P = 0.02) and water content was decreased (8.6%; P < 0.01) whereas in RV, wall thickness was decreased (26%; P = 0.04) and water content was similar in EIHS compared to TN. We also discovered ventricle-specific biochemical changes such that in RV EIHS increased heat shock proteins, decreased AMPK and AKT signaling, decreased activation of mTOR (35%; P < 0.05), and increased expression of proteins that participate in autophagy. In LV, heat shock proteins, AMPK and AKT signaling, activation of mTOR, and autophagy-related proteins were largely similar between groups. Biomarkers suggest EIHS-mediated reductions in kidney function. These data demonstrate EIHS causes ventricular-dependent changes and may undermine cardiac health, energy homeostasis, and function.
Collapse
|
17
|
Bissoli I, D’Adamo S, Pignatti C, Agnetti G, Flamigni F, Cetrullo S. Induced pluripotent stem cell-based models: Are we ready for that heart in a dish? Front Cell Dev Biol 2023; 11:1129263. [PMID: 36743420 PMCID: PMC9892938 DOI: 10.3389/fcell.2023.1129263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Affiliation(s)
- Irene Bissoli
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Stefania D’Adamo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carla Pignatti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giulio Agnetti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
- Center for Research on Cardiac Intermediate Filaments, Johns Hopkins University, Baltimore, MD, United States
| | - Flavio Flamigni
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| | - Silvia Cetrullo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| |
Collapse
|
18
|
Sazzad F, Kollengode R, Beverly CLX, Kiat TY, Ganesh G, Kofidis T. Preclinical Large Animal In-Vivo Experiments for Surgically Implanted Atrioventricular Valve: Reappraisal and Systematic Review. Curr Cardiol Rev 2023; 19:e170622206130. [PMID: 35718960 PMCID: PMC10201874 DOI: 10.2174/1573403x18666220617115216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The development of atrioventricular bioprosthesis has witnessed an increasing drive toward clinical translation over the last few decades. A significant challenge in the clinical translation of an atrioventricular bioprosthesis from bench to bedside is the appropriate choice of a large animal model to test the safety and effectiveness of the device. METHODS We conducted a systematic review of pre-clinical in vivo studies that would enable us to synthesize a recommended framework. PRISMA (Preferred Reporting Items for Systematic Reviews and MetaAnalyses) guidelines were followed to identify and extract relevant articles. RESULTS Sheep was the most common choice of animal, with nine out of the 12 included studies being conducted on sheep. There were acute and chronic studies based on our search criteria. An average of ~20 and 5 animals were used for chronic and acute studies. One out of three acute studies and eight out of nine chronic studies were on stented heart valve bioprosthesis. All analyses were conducted on the implantation of atrioventricular valves with trileaflet, except for one chronic study on unileaflet valves and one chronic and acute study on bileaflet valves. CONCLUSION Understanding the variance in past pre-clinical study designs may increase the appropriate utilization of large animal models. This synthesized evidence provides a pre-clinical in vivo studies framework for future research on an atrioventricular bioprosthesis.
Collapse
Affiliation(s)
- Faizus Sazzad
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Center, Kent Ridge, Singapore
| | - Ramanathan Kollengode
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Center, Kent Ridge, Singapore
| | - Chan Li Xuan Beverly
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Tan Ying Kiat
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Geetha Ganesh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Theo Kofidis
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Center, Kent Ridge, Singapore
| |
Collapse
|
19
|
Pan Z, Liang P. Human-Induced Pluripotent Stem Cell-Based Differentiation of Cardiomyocyte Subtypes for Drug Discovery and Cell Therapy. Handb Exp Pharmacol 2023; 281:209-233. [PMID: 37421443 DOI: 10.1007/164_2023_663] [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] [Indexed: 07/10/2023]
Abstract
Drug attrition rates have increased over the past few years, accompanied with growing costs for the pharmaceutical industry and consumers. Lack of in vitro models connecting the results of toxicity screening assays with clinical outcomes accounts for this high attrition rate. The emergence of cardiomyocytes derived from human pluripotent stem cells provides an amenable source of cells for disease modeling, drug discovery, and cardiotoxicity screening. Functionally similar to to embryonic stem cells, but with fewer ethical concerns, induced pluripotent stem cells (iPSCs) can recapitulate patient-specific genetic backgrounds, which would be a huge revolution for personalized medicine. The generated iPSC-derived cardiomyocytes (iPSC-CMs) represent different subtypes including ventricular-, atrial-, and nodal-like cardiomyocytes. Purifying these subtypes for chamber-specific drug screening presents opportunities and challenges. In this chapter, we discuss the strategies for the purification of iPSC-CMs, the use of iPSC-CMs for drug discovery and cardiotoxicity test, and the current limitations of iPSC-CMs that should be overcome for wider and more precise cardiovascular applications.
Collapse
Affiliation(s)
- Ziwei Pan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
20
|
van der Velden J, Asselbergs FW, Bakkers J, Batkai S, Bertrand L, Bezzina CR, Bot I, Brundel BJJM, Carrier L, Chamuleau S, Ciccarelli M, Dawson D, Davidson SM, Dendorfer A, Duncker DJ, Eschenhagen T, Fabritz L, Falcão-Pires I, Ferdinandy P, Giacca M, Girao H, Gollmann-Tepeköylü C, Gyongyosi M, Guzik TJ, Hamdani N, Heymans S, Hilfiker A, Hilfiker-Kleiner D, Hoekstra AG, Hulot JS, Kuster DWD, van Laake LW, Lecour S, Leiner T, Linke WA, Lumens J, Lutgens E, Madonna R, Maegdefessel L, Mayr M, van der Meer P, Passier R, Perbellini F, Perrino C, Pesce M, Priori S, Remme CA, Rosenhahn B, Schotten U, Schulz R, Sipido KR, Sluijter JPG, van Steenbeek F, Steffens S, Terracciano CM, Tocchetti CG, Vlasman P, Yeung KK, Zacchigna S, Zwaagman D, Thum T. Animal models and animal-free innovations for cardiovascular research: current status and routes to be explored. Consensus document of the ESC Working Group on Myocardial Function and the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2022; 118:3016-3051. [PMID: 34999816 PMCID: PMC9732557 DOI: 10.1093/cvr/cvab370] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases represent a major cause of morbidity and mortality, necessitating research to improve diagnostics, and to discover and test novel preventive and curative therapies, all of which warrant experimental models that recapitulate human disease. The translation of basic science results to clinical practice is a challenging task, in particular for complex conditions such as cardiovascular diseases, which often result from multiple risk factors and comorbidities. This difficulty might lead some individuals to question the value of animal research, citing the translational 'valley of death', which largely reflects the fact that studies in rodents are difficult to translate to humans. This is also influenced by the fact that new, human-derived in vitro models can recapitulate aspects of disease processes. However, it would be a mistake to think that animal models do not represent a vital step in the translational pathway as they do provide important pathophysiological insights into disease mechanisms particularly on an organ and systemic level. While stem cell-derived human models have the potential to become key in testing toxicity and effectiveness of new drugs, we need to be realistic, and carefully validate all new human-like disease models. In this position paper, we highlight recent advances in trying to reduce the number of animals for cardiovascular research ranging from stem cell-derived models to in situ modelling of heart properties, bioinformatic models based on large datasets, and state-of-the-art animal models, which show clinically relevant characteristics observed in patients with a cardiovascular disease. We aim to provide a guide to help researchers in their experimental design to translate bench findings to clinical routine taking the replacement, reduction, and refinement (3R) as a guiding concept.
Collapse
Grants
- R01 HL150359 NHLBI NIH HHS
- RG/16/14/32397 British Heart Foundation
- FS/18/37/33642 British Heart Foundation
- PG/17/64/33205 British Heart Foundation
- PG/15/88/31780 British Heart Foundation
- FS/RTF/20/30009, NH/19/1/34595, PG/18/35/33786, CS/17/4/32960, PG/15/88/31780, and PG/17/64/33205 British Heart Foundation
- NC/T001488/1 National Centre for the Replacement, Refinement and Reduction of Animals in Research
- PG/18/44/33790 British Heart Foundation
- CH/16/3/32406 British Heart Foundation
- FS/RTF/20/30009 British Heart Foundation
- NWO-ZonMW
- ZonMW and Heart Foundation for the translational research program
- Dutch Cardiovascular Alliance (DCVA)
- Leducq Foundation
- Dutch Research Council
- Association of Collaborating Health Foundations (SGF)
- UCL Hospitals NIHR Biomedical Research Centre, and the DCVA
- Netherlands CardioVascular Research Initiative CVON
- Stichting Hartekind and the Dutch Research Counsel (NWO) (OCENW.GROOT.2019.029)
- National Fund for Scientific Research, Belgium and Action de Recherche Concertée de la Communauté Wallonie-Bruxelles, Belgium
- Netherlands CardioVascular Research Initiative CVON (PREDICT2 and CONCOR-genes projects), the Leducq Foundation
- ERA PerMed (PROCEED study)
- Netherlands Cardiovascular Research Initiative
- Dutch Heart Foundation
- German Centre of Cardiovascular Research (DZHH)
- Chest Heart and Stroke Scotland
- Tenovus Scotland
- Friends of Anchor and Grampian NHS-Endowments
- National Institute for Health Research University College London Hospitals Biomedical Research Centre
- German Centre for Cardiovascular Research
- European Research Council (ERC-AG IndivuHeart), the Deutsche Forschungsgemeinschaft
- European Union Horizon 2020 (REANIMA and TRAINHEART)
- German Ministry of Education and Research (BMBF)
- Centre for Cardiovascular Research (DZHK)
- European Union Horizon 2020
- DFG
- National Research, Development and Innovation Office of Hungary
- Research Excellence Program—TKP; National Heart Program
- Austrian Science Fund
- European Union Commission’s Seventh Framework programme
- CVON2016-Early HFPEF
- CVON She-PREDICTS
- CVON Arena-PRIME
- European Union’s Horizon 2020 research and innovation programme
- Deutsche Forschungsgemeinschaft
- Volkswagenstiftung
- French National Research Agency
- ERA-Net-CVD
- Fédération Française de Cardiologie, the Fondation pour la Recherche Médicale
- French PIA Project
- University Research Federation against heart failure
- Netherlands Heart Foundation
- Dekker Senior Clinical Scientist
- Health Holland TKI-LSH
- TUe/UMCU/UU Alliance Fund
- south African National Foundation
- Cancer Association of South Africa and Winetech
- Netherlands Heart Foundation/Applied & Engineering Sciences
- Dutch Technology Foundation
- Pie Medical Imaging
- Netherlands Organisation for Scientific Research
- Dr. Dekker Program
- Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation
- Dutch Federation of University Medical Centres
- Netherlands Organization for Health Research and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project
- Netherlands Organization for Scientific Research (NWO) (VICI grant); the European Research Council
- Incyte s.r.l. and from Ministero dell’Istruzione, Università e Ricerca Scientifica
- German Center for Cardiovascular Research (Junior Research Group & Translational Research Project), the European Research Council (ERC Starting Grant NORVAS),
- Swedish Heart-Lung-Foundation
- Swedish Research Council
- National Institutes of Health
- Bavarian State Ministry of Health and Care through the research project DigiMed Bayern
- ERC
- ERA-CVD
- Dutch Heart Foundation, ZonMw
- the NWO Gravitation project
- Ministero dell'Istruzione, Università e Ricerca Scientifica
- Regione Lombardia
- Netherlands Organisation for Health Research and Development
- ITN Network Personalize AF: Personalized Therapies for Atrial Fibrillation: a translational network
- MAESTRIA: Machine Learning Artificial Intelligence Early Detection Stroke Atrial Fibrillation
- REPAIR: Restoring cardiac mechanical function by polymeric artificial muscular tissue
- Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)
- European Union H2020 program to the project TECHNOBEAT
- EVICARE
- BRAV3
- ZonMw
- German Centre for Cardiovascular Research (DZHK)
- British Heart Foundation Centre for Cardiac Regeneration
- British Heart Foundation studentship
- NC3Rs
- Interreg ITA-AUS project InCARDIO
- Italian Association for Cancer Research
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, UK
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sandor Batkai
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Luc Bertrand
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Connie R Bezzina
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Ilze Bot
- Heart Center, Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Steven Chamuleau
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Odontology, University of Salerno, Fisciano (SA), Italy
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- University Center of Cardiovascular Sciences and Department of Cardiology, University Heart Center Hamburg, Germany and Institute of Cardiovascular Sciences, University of Birmingham, UK
| | - Ines Falcão-Pires
- UnIC - Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Portugal
| | - Péter Ferdinandy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Henrique Girao
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, Faculty of Medicine, Coimbra, Portugal
- Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | | | - Mariann Gyongyosi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Tomasz J Guzik
- Instutute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Nazha Hamdani
- Division Cardiology, Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Andres Hilfiker
- Department for Cardiothoracic, Transplant, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Comprehensive Cancer Centre, Philipps-Universität Marburg, Germany
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Sébastien Hulot
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Linda W van Laake
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Tim Leiner
- Department of Radiology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27B, 48149 Muenster, Germany
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, 56124 Pisa, Italy
- Department of Internal Medicine, Cardiology Division, University of Texas Medical School in Houston, Houston, TX, USA
| | - Lars Maegdefessel
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500AE Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Filippo Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro cardiologico Monzino, IRCCS, Milan, Italy
| | - Silvia Priori
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, Pavia, Italy
- University of Pavia, Pavia, Italy
| | - Carol Ann Remme
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Bodo Rosenhahn
- Institute for information Processing, Leibniz University of Hanover, 30167 Hannover, Germany
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karin R Sipido
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, Regenerative Medicine Center Utrecht, Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Steenbeek
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center for Clinical and Translational Research (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Patricia Vlasman
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Amsterdam UMC, Vrije Universiteit, Surgery, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Dayenne Zwaagman
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Thomas Thum
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
21
|
Marynowska M, Herosimczyk A, Lepczyński A, Barszcz M, Konopka A, Dunisławska A, Ożgo M. Gene and Protein Accumulation Changes Evoked in Porcine Aorta in Response to Feeding with Two Various Fructan Sources. Animals (Basel) 2022; 12:3147. [PMID: 36428375 PMCID: PMC9687048 DOI: 10.3390/ani12223147] [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: 09/29/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, two different ITFs sources were incorporated into a cereal-based diet to evaluate possible aortic protein and gene changes in nursery pigs. The animals were fed two different experimental diets from the 10th day of life, supplemented with either 4% of dried chicory root (CR) or with 2% of native inulin (IN). After a 40-day dietary intervention trial, pigs were sacrificed at day 50 and the aortas were harvested. Our data indicate that dietary ITFs have the potential to influence several structural and physiological changes that are reflected both in the mRNA and protein levels in porcine aorta. In contrast to our hypothesis, we could not show any beneficial effects of a CR diet on vascular functions. The direction of changes of several proteins and genes may indicate disrupted ECM turnover (COL6A1 and COL6A2, MMP2, TIMP3, EFEMP1), increased inflammation and lipid accumulation (FFAR2), as well as decreased activity of endothelial nitric oxide synthase (TXNDC5, ORM1). On the other hand, the IN diet may counteract a highly pro-oxidant environment through the endothelin-NO axis (CALR, TCP1, HSP8, PDIA3, RCN2), fibrinolytic activity (ANXA2), anti-atherogenic (CAVIN-1) and anti-calcification (LMNA) properties, thus contributing to the maintenance of vascular homeostasis.
Collapse
Affiliation(s)
- Marta Marynowska
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland
| | - Agnieszka Herosimczyk
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland
| | - Adam Lepczyński
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland
| | - Marcin Barszcz
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland
| | - Adrianna Konopka
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland
| | - Aleksandra Dunisławska
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Małgorzata Ożgo
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland
| |
Collapse
|
22
|
Mandour AS, Samir H, Yoshida T, Matsuura K, Hamabe L, Shimada K, Abdelmageed HA, Elbadawy M, Uemura A, Takahashi K, Watanabe G, Tanaka R. Novel color M-mode echocardiography for non-invasive assessment of the intraventricular pressure in goats: Feasibility, repeatability, and the effect of sedation. Front Vet Sci 2022; 9:935437. [PMID: 36277071 PMCID: PMC9582648 DOI: 10.3389/fvets.2022.935437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
Background The intraventricular pressure difference (IVPD) and intraventricular pressure gradients (IVPG), estimated from color M-mode echocardiography (CMME) of the transmitral flow, have been introduced as novel indices for the evaluation of heart functions. Until now, no study demonstrated the feasibility of the CMME approach to measure IVPD and IVPG in any farm animals. The aim of this study was to assess the feasibility and repeatability of CMME-derived IVPD and IVPG variables in goats and explore the effect of sedation on the measured variables. Materials and methods Sixteen male Shiba goats were included in this study and underwent conventional echocardiography. Eight goats were used in the repeatability of IVPD/IVPG variables. Another eight goats were used to evaluate the effect of sedation by xylazine on IVPD/IVPG measurements. CMME between the base and the apex of the left ventricle was carried out. The IVPD and IVPG were analyzed using in-house code software. The IVPD and IVPG were expressed as total, basal, mid-to-apical, mid, and apical segments. Data analysis including the imaging quality score (IQS), repeatability, variability, intraclass correlation coefficient (ICC), as well as the effect size of sedation on the measured variables was calculated. Results IVPD and IVPG variables from CMME were feasible in all goats. Low to moderate variability of IVPD and IVPG variables was observed (CV 95% <25%) except for the apical IVPD and apical IVPG. The IVPD/IVPG measurements were repeatable without a significant effect of animal or time on the obtained measurements. The overall ICC was higher than 0.75 in all variables except for the apical segment. Xylazine administration reduced the total, basal, and mid parts of IVPD and IVPG with a large effect size (biserial ranked correlation; rc > 0.8). Conclusion We reported, for the first time, IVPD and IVPG measurements by CMME in goats. The assessment of IVPD and IVPG by CMME is feasible in goats which can be evaluated in further cardiovascular or pharmacological studies in this species.
Collapse
Affiliation(s)
- Ahmed S. Mandour
- Department of Veterinary Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt,Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan,*Correspondence: Ahmed S. Mandour
| | - Haney Samir
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Tomohiko Yoshida
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Katsuhiro Matsuura
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Lina Hamabe
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazumi Shimada
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hend A. Abdelmageed
- Ismailia Laboratory, Animal Health Research Institute, Agriculture Research Center, First District, Ismailia, Egypt
| | - Mohamed Elbadawy
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt,Laboratory of Pharmacology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Akiko Uemura
- Division of Veterinary Research, Department of Veterinary Surgery, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ken Takahashi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan,Ryou Tanaka
| |
Collapse
|
23
|
Metabolomic Profiling of End-Stage Heart Failure Secondary to Chronic Chagas Cardiomyopathy. Int J Mol Sci 2022; 23:ijms231810456. [PMID: 36142367 PMCID: PMC9499603 DOI: 10.3390/ijms231810456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is the most frequent and severe clinical form of chronic Chagas disease, representing one of the leading causes of morbidity and mortality in Latin America, and a growing global public health problem. There is currently no approved treatment for CCC; however, omics technologies have enabled significant progress to be made in the search for new therapeutic targets. The metabolic alterations associated with pathogenic mechanisms of CCC and their relationship to cellular and immunopathogenic processes in cardiac tissue remain largely unknown. This exploratory study aimed to evaluate the potential underlying pathogenic mechanisms in the failing myocardium of patients with end-stage heart failure (ESHF) secondary to CCC by applying an untargeted metabolomic profiling approach. Cardiac tissue samples from the left ventricle of patients with ESHF of CCC etiology (n = 7) and healthy donors (n = 7) were analyzed using liquid chromatography-mass spectrometry. Metabolite profiles showed altered branched-chain amino acid and acylcarnitine levels, decreased fatty acid uptake and oxidation, increased activity of the pentose phosphate pathway, dysregulation of the TCA cycle, and alterations in critical cellular antioxidant systems. These findings suggest processes of energy deficit, alterations in substrate availability, and enhanced production of reactive oxygen species in the affected myocardium. This profile potentially contributes to the development and maintenance of a chronic inflammatory state that leads to progression and severity of CCC. Further studies involving larger sample sizes and comparisons with heart failure patients without CCC are needed to validate these results, opening an avenue to investigate new therapeutic approaches for the treatment and prevention of progression of this unique and severe cardiomyopathy.
Collapse
|
24
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|
25
|
The Sheep as a Large Animal Model for the Investigation and Treatment of Human Disorders. BIOLOGY 2022; 11:biology11091251. [PMID: 36138730 PMCID: PMC9495394 DOI: 10.3390/biology11091251] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/19/2022]
Abstract
Simple Summary We review the value of large animal models for improving the translation of biomedical research for human application, focusing primarily on sheep. Abstract An essential aim of biomedical research is to translate basic science information obtained from preclinical research using small and large animal models into clinical practice for the benefit of humans. Research on rodent models has enhanced our understanding of complex pathophysiology, thus providing potential translational pathways. However, the success of translating drugs from pre-clinical to clinical therapy has been poor, partly due to the choice of experimental model. The sheep model, in particular, is being increasingly applied to the field of biomedical research and is arguably one of the most influential models of human organ systems. It has provided essential tools and insights into cardiovascular disorder, orthopaedic examination, reproduction, gene therapy, and new insights into neurodegenerative research. Unlike the widely adopted rodent model, the use of the sheep model has an advantage over improving neuroscientific translation, in particular due to its large body size, gyrencephalic brain, long lifespan, more extended gestation period, and similarities in neuroanatomical structures to humans. This review aims to summarise the current status of sheep to model various human diseases and enable researchers to make informed decisions when considering sheep as a human biomedical model.
Collapse
|
26
|
Dora KA, Lin J, Borysova L, Beleznai T, Taggart M, Ascione R, Garland C. Signaling and structures underpinning conducted vasodilation in human and porcine intramyocardial coronary arteries. Front Cardiovasc Med 2022; 9:980628. [PMID: 36035957 PMCID: PMC9411971 DOI: 10.3389/fcvm.2022.980628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 01/28/2023] Open
Abstract
Background Adequate blood flow into coronary micro-arteries is essential for myocardial function. Here we assess the mechanisms responsible for amplifying blood flow into myogenically-contracting human and porcine intramyocardial micro-arteries ex vivo using endothelium-dependent and -independent vasodilators. Methods Human and porcine atrial and ventricular small intramyocardial coronary arteries (IMCAs) were studied with pressure myography and imaged using confocal microscopy and serial section/3-D reconstruction EM. Results 3D rendered ultrastructure images of human right atrial (RA-) IMCAs revealed extensive homo-and hetero-cellular contacts, including to longitudinally-arranged smooth muscle cells (l-SMCs) found between the endothelial cells (ECs) and radially-arranged medial SMCs (r-SMCs). Local and conducted vasodilatation followed focal application of bradykinin in both human and porcine RA-IMCAs, and relied on hyperpolarization of SMCs, but not nitric oxide. Bradykinin initiated asynchronous oscillations in endothelial cell Ca2+ in pressurized RA-IMCAs and, as previously shown in human RA-IMCAs, hyperpolarized porcine arteries. Immunolabelling showed small- and intermediate-conductance Ca2+-activated K+ channels (KCa) present in the endothelium of both species, and concentration-dependent vasodilation to bradykinin followed activation of these KCa channels. Extensive electrical coupling was demonstrated between r-SMCs and l-SMCs, providing an additional pathway to facilitate the well-established myoendothelial coupling. Conducted dilation was still evident in a human RA-IMCA with poor myogenic tone, and heterocellular contacts were visible in the 3D reconstructed artery. Hyperpolarization and conducted vasodilation was also observed to adenosine which, in contrast to bradykinin, was sensitive to combined block of ATP-sensitive (KATP) and inwardly rectifying (KIR) K+ channels. Conclusions These data extend our understanding of the mechanisms that coordinate human coronary microvascular blood flow and the mechanistic overlap with porcine IMCAs. The unusual presence of l-SMCs provides an additional pathway for rapid intercellular signaling between cells of the coronary artery wall. Local and conducted vasodilation follow hyperpolarization of the ECs or SMCs, and contact-coupling between l-SMCs and r-SMCs likely facilitates this vasodilation.
Collapse
Affiliation(s)
- Kim A Dora
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - JinHeng Lin
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Lyudmyla Borysova
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Timea Beleznai
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Michael Taggart
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raimondo Ascione
- Bristol Heart Institute and Translational Biomedical Research Centre, University of Bristol, Bristol, United Kingdom
| | - Christopher Garland
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Martin SC, Hauser N, Renaldo AC, Lane M, Jordan JE, Qadri HI, Mouser N, Rahbar E, Williams TK, Neff LP. Unmasking the Confounder: The Inherent Physiologic Variability of Swine During an Automated Experimental Model of Ischemia-Reperfusion Injury. Am Surg 2022; 88:1838-1844. [PMID: 35392677 DOI: 10.1177/00031348221084967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND We sought to determine the magnitude of the inherent inter-animal physiologic variability by automating a porcine Resuscitative Endovascular Balloon Occlusion of the Aorta (REBOA) protocol to minimize external influences that might alter physiology and confound experimental results. METHODS Swine (n = 42) underwent a controlled 30% blood volume hemorrhage followed by 30 minutes of REBOA (ie, ischemic phase). The animals were weaned from REBOA autonomously over 15 minutes, beginning the reperfusion phase, while continuing to provide partial flow balloon support to maintain a target proximal mean arterial pressure (pMAP) of 65 mmHg. Simultaneously, shed blood was re-transfused as part of the resuscitation efforts. Physiologic data were continuously recorded, and serum samples were serially collected. Baseline characteristics, variance in vital signs, and 8-isoprostane levels were quantified during hemorrhage, REBOA, and reperfusion phases. RESULTS There was no significant difference in baseline physiology across animals (P > .05). Hemodynamic variability was highest for pMAP during the ischemic phase (P = .001) and for distal mean arterial pressure (dMAP) during the weaning/reperfusion phase (P = .001). The latter finding indicated the variable physiologic response to ischemia-reperfusion injury, as the automated balloon support required by each animal to maintain pMAP was highly variable. Circulating 8-isoprostane variance was significantly higher following the start of reperfusion compared to baseline levels (P = .001). DISCUSSION Despite subjecting animals to a highly consistent ischemia-reperfusion injury through automation, we noted significant variability in the hemodynamic and biochemical response. These findings illustrate the inherent physiologic variability and potential limitations of porcine large animal models for the study of shock.
Collapse
Affiliation(s)
| | - Nathaniel Hauser
- Department of Biomedical Engineering, 12279Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Antonio C Renaldo
- Department of Biomedical Engineering, 12279Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Magan Lane
- Department of Vascular and Endovascular Surgery, 12280Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - James E Jordan
- Department of Cardiothoracic Surgery, 12280Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Hisham I Qadri
- 12279Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Elaheh Rahbar
- Department of Biomedical Engineering, 12279Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Timothy K Williams
- Department of Vascular and Endovascular Surgery, 12280Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Lucas P Neff
- Department of General Surgery, 12280Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| |
Collapse
|
28
|
Maselli D, Matos RS, Johnson RD, Martella D, Caprettini V, Chiappini C, Camelliti P, Campagnolo P. Porcine Organotypic Epicardial Slice Protocol: A Tool for the Study of Epicardium in Cardiovascular Research. Front Cardiovasc Med 2022; 9:920013. [PMID: 35924218 PMCID: PMC9339655 DOI: 10.3389/fcvm.2022.920013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The epicardium has recently gained interest in the cardiovascular field due to its capacity to support heart regeneration after ischemic injury. Models to study the epicardium of large animals in vitro are limited and mainly based on epicardial cell isolation/differentiation from stem cells, followed by 2D cells culture. In this method paper, we describe the procedure to obtain and culture 3D organotypic heart slices presenting an intact epicardium, as a novel model to study the epicardial physiology and activation. Epicardial slices are obtained from porcine hearts using a high-precision vibratome and retain a healthy epicardial layer embedded in its native extracellular environment and connected with other cardiac cells (cardiomyocytes, fibroblasts, vascular cells etc.). Epicardial slices can be cultured for 72 h, providing an ideal model for studying the epicardium physiology or perform pharmacological interventions/gene therapy approaches. We also report on methods to assesses the viability and composition of the epicardial slices, and evaluate their architecture in 3D through tissue decoloration. Finally, we present a potential application for a nanomaterial-based gene transfer method for tracking of epicardial cells within the slice. Crucially, given the similarity in morphology and physiology of porcine heart with its human counterpart, our system provides a platform for translational research while providing a clinically relevant and ethical alternative to the use of small animals in this type of research.
Collapse
Affiliation(s)
- Davide Maselli
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Rolando S. Matos
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Robert D. Johnson
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Davide Martella
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Valeria Caprettini
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Ciro Chiappini
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Patrizia Camelliti
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Paola Campagnolo
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
29
|
Liu Z, Andraska E, Akinbode D, Mars W, Alvidrez RIM. LRP1 in the Vascular Wall. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Gaidulis G, Suresh KS, Xu D, Padala M. Patient-Specific Three-Dimensional Ultrasound Derived Computational Modeling of the Mitral Valve. Ann Biomed Eng 2022; 50:847-859. [PMID: 35380321 PMCID: PMC10826907 DOI: 10.1007/s10439-022-02960-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/27/2022] [Indexed: 11/01/2022]
Abstract
Several new techniques to repair the mitral valve affected by functional mitral regurgitation are in development. However, due to the heterogeneity of valve lesions between patients, predicting the outcomes of novel treatment approaches is challenging. We present a patient-specific, 3D ultrasound-derived computational model of the mitral valve for procedure planning, that faithfully mimics the pathological valve dynamics. 3D ultrasound images were obtained in three pigs induced with heart failure and which developed functional mitral regurgitation. For each case, images were segmented, and finite element model of mitral valve was constructed. Annular and papillary muscle dynamics were extracted and imposed as kinematic boundary conditions, and the chordae were pre-strained to induce valve tethering. Valve closure was simulated by applying physiologic transvalvular pressure on the leaflets. Agreement between simulation results and truth datasets was confirmed, with accurate location of regurgitation jets and coaptation defects. Inclusion of kinematic patient-specific boundary conditions was necessary to achieve these results, whereas use of idealized boundary conditions deviated from the truth dataset. Due to the impact of boundary conditions on the model, the effect of repair strategies on valve closure varied as well, indicating that our approach of using patient-specific boundary conditions for mitral valve modeling is valid.
Collapse
Affiliation(s)
- Gediminas Gaidulis
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, 380B Northyards Blvd NW, Atlanta, GA, 30313, USA
- Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Kirthana Sreerangathama Suresh
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, 380B Northyards Blvd NW, Atlanta, GA, 30313, USA
| | - Dongyang Xu
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, 380B Northyards Blvd NW, Atlanta, GA, 30313, USA
| | - Muralidhar Padala
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, 380B Northyards Blvd NW, Atlanta, GA, 30313, USA.
- Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
31
|
Cardiovascular Diseases in the Digital Health Era: A Translational Approach from the Lab to the Clinic. BIOTECH 2022; 11:biotech11030023. [PMID: 35892928 PMCID: PMC9326743 DOI: 10.3390/biotech11030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/19/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Translational science has been introduced as the nexus among the scientific and the clinical field, which allows researchers to provide and demonstrate that the evidence-based research can connect the gaps present between basic and clinical levels. This type of research has played a major role in the field of cardiovascular diseases, where the main objective has been to identify and transfer potential treatments identified at preclinical stages into clinical practice. This transfer has been enhanced by the intromission of digital health solutions into both basic research and clinical scenarios. This review aimed to identify and summarize the most important translational advances in the last years in the cardiovascular field together with the potential challenges that still remain in basic research, clinical scenarios, and regulatory agencies.
Collapse
|
32
|
Dora KA, Borysova L, Ye X, Powell C, Beleznai TZ, Stanley CP, Bruno VD, Starborg T, Johnson E, Pielach A, Taggart M, Smart N, Ascione R. Human coronary microvascular contractile dysfunction associates with viable synthetic smooth muscle cells. Cardiovasc Res 2022; 118:1978-1992. [PMID: 34173824 PMCID: PMC9239576 DOI: 10.1093/cvr/cvab218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
AIMS Coronary microvascular smooth muscle cells (SMCs) respond to luminal pressure by developing myogenic tone (MT), a process integral to the regulation of microvascular perfusion. The cellular mechanisms underlying poor myogenic reactivity in patients with heart valve disease are unknown and form the focus of this study. METHODS AND RESULTS Intramyocardial coronary micro-arteries (IMCAs) isolated from human and pig right atrial (RA) appendage and left ventricular (LV) biopsies were studied using pressure myography combined with confocal microscopy. All RA- and LV-IMCAs from organ donors and pigs developed circa 25% MT. In contrast, 44% of human RA-IMCAs from 88 patients with heart valve disease had poor (<10%) MT yet retained cell viability and an ability to raise cytoplasmic Ca2+ in response to vasoconstrictor agents. Comparing across human heart chambers and species, we found that based on patient medical history and six tests, the strongest predictor of poor MT in IMCAs was increased expression of the synthetic marker caldesmon relative to the contractile marker SM-myosin heavy chain. In addition, high resolution imaging revealed a distinct layer of longitudinally aligned SMCs between ECs and radial SMCs, and we show poor MT was associated with disruptions in these cellular alignments. CONCLUSION These data demonstrate the first use of atrial and ventricular biopsies from patients and pigs to reveal that impaired coronary MT reflects a switch of viable SMCs towards a synthetic phenotype, rather than a loss of SMC viability. These arteries represent a model for further studies of coronary microvascular contractile dysfunction.
Collapse
Affiliation(s)
- Kim A Dora
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Lyudmyla Borysova
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Xi Ye
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Chloe Powell
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Timea Z Beleznai
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Christopher P Stanley
- The Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Vito D Bruno
- Bristol Heart Institute and Translational Biomedical Research Centre, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Tobias Starborg
- Division of Cell Matrix Biology and Regenerative Medicine School of Biological Sciences Faculty of Biology, Medical and Health Sciences, University of Manchester, B.3016 Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Anna Pielach
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Michael Taggart
- Biosciences Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Raimondo Ascione
- Bristol Heart Institute and Translational Biomedical Research Centre, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| |
Collapse
|
33
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
34
|
Li SJ, Cheng WL, Kao YH, Chung CC, Trang NN, Chen YJ. Melatonin Inhibits NF-κB/CREB/Runx2 Signaling and Alleviates Aortic Valve Calcification. Front Cardiovasc Med 2022; 9:885293. [PMID: 35795373 PMCID: PMC9251177 DOI: 10.3389/fcvm.2022.885293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is linked to high mortality. Melatonin inhibits nuclear factor-kappa B (NF-κB)/cyclic AMP response element-binding protein (CREB), contributing to CAVD progression. This study determined the role of melatonin/MT1/MT2 signaling in valvular interstitial cell (VIC) calcification. Western blotting and Alizarin red staining were used to analyze NF-κB/CREB/runt-related transcription factor 2 (Runx2) signaling in porcine VICs treated with an osteogenic (OST) medium without (control) or with melatonin for 5 days. Chromatin immunoprecipitation (ChIP) assay was used to analyze NF-κB's transcription regulation of NF-κB on the Runx2 promoter. OST medium-treated VICs exhibited a greater expression of NF-κB, CREB, and Runx2 than control VICs. Melatonin treatment downregulated the effects of the OST medium and reduced VIC calcification. The MT1/MT2 antagonist (Luzindole) and MT1 receptor neutralized antibody blocked the anticalcification effect of melatonin, but an MT2-specific inhibitor (4-P-PDOT) did not. Besides, the NF-κB inhibitor (SC75741) reduced OST medium-induced VIC calcification to a similar extent to melatonin at 10 nmol/L. The ChIP assay demonstrated that melatonin attenuated OST media increased NF-κB binding activity to the promoter region of Runx2. Activation of the melatonin/MT1-axis significantly reduced VIC calcification by targeting the NF-κB/CREB/Runx2 pathway. Targeting melatonin/MT1 signaling may be a potential therapeutic strategy for CAVD.
Collapse
Affiliation(s)
- Shao-Jung Li
- Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Wan-Li Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chih Chung
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Yi-Jen Chen
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Yi-Jen Chen
| |
Collapse
|
35
|
Transcriptional and Epigenetic Factors Associated with Early Thrombosis of Femoral Artery Involved in Arteriovenous Fistula. Proteomes 2022; 10:proteomes10020014. [PMID: 35645372 PMCID: PMC9149803 DOI: 10.3390/proteomes10020014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Arteriovenous fistulas (AVFs), created for hemodialysis in end-stage renal disease patients, mature through the outward remodeling of the outflow vein. However, early thrombosis and chronic inflammation are detrimental to the process of AVF maturation and precipitate AVF maturation failure. For the successful remodeling of the outflow vein, blood flow through the fistula is essential, but early arterial thrombosis attenuates this blood flow, and the vessels become thrombosed and stenosed, leading to AVF failure. The altered expression of various proteins involved in maintaining vessel patency or thrombosis is regulated by genes of which the expression is regulated by transcription factors and microRNAs. In this study, using thrombosed and stenosed arteries following AVF creation, we delineated transcription factors and microRNAs associated with differentially expressed genes in bulk RNA sequencing data using upstream and causal network analysis. We observed changes in many transcription factors and microRNAs that are involved in angiogenesis; vascular smooth muscle cell proliferation, migration, and phenotypic changes; endothelial cell function; hypoxia; oxidative stress; vessel remodeling; immune responses; and inflammation. These factors and microRNAs play a critical role in the underlying molecular mechanisms in AVF maturation. We also observed epigenetic factors involved in gene regulation associated with these molecular mechanisms. The results of this study indicate the importance of investigating the transcriptional and epigenetic regulation of AVF maturation and maturation failure and targeting factors precipitating early thrombosis and stenosis.
Collapse
|
36
|
Awad K, Sayed A, Banach M. Coenzyme Q10 Reduces Infarct Size in Animal Models of Myocardial Ischemia-Reperfusion Injury: A Meta-Analysis and Summary of Underlying Mechanisms. Front Cardiovasc Med 2022; 9:857364. [PMID: 35498032 PMCID: PMC9053645 DOI: 10.3389/fcvm.2022.857364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/15/2022] [Indexed: 01/14/2023] Open
Abstract
Objective Effective interventions that might limit myocardial ischemia-reperfusion (I/R) injury are still lacking. Coenzyme Q10 (CoQ10) may exert cardioprotective actions that reduce myocardial I/R injury. We conducted this meta-analysis to assess the potential cardioprotective effect of CoQ10 in animal models of myocardial I/R injury. Methods We searched PubMed and Embase databases from inception to February 2022 to identify animal studies that compared the effect of CoQ10 with vehicle treatment or no treatment on myocardial infarct size in models of myocardial I/R injury. Means and standard deviations of the infarct size measurements were pooled as the weighted mean difference with 95% confidence interval (CI) using the random-effects model. Subgroup analyses were also conducted according to animals' species, models' type, and reperfusion time. Results Six animal studies (4 in vivo and 2 ex vivo) with 116 animals were included. Pooled analysis suggested that CoQ10 significantly reduced myocardial infarct size by −11.36% (95% CI: −16.82, −5.90, p < 0.0001, I2 = 94%) compared with the control group. The significance of the pooled effect estimate was maintained in rats, Hartley guinea pigs, and Yorkshire pigs. However, it became insignificant in the subgroup of rabbits −5.29% (95% CI: −27.83, 17.26; I2 = 87%). Furthermore, CoQ10 significantly reduced the myocardial infarct size regardless of model type (either in vivo or ex vivo) and reperfusion time (either ≤ 4 h or >4 h). Conclusion Coenzyme Q10 significantly decreased myocardial infarct size by 11.36% compared with the control group in animal models of myocardial I/R injury. This beneficial action was retained regardless of model type and reperfusion time.
Collapse
Affiliation(s)
- Kamal Awad
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Zagazig University Hospitals, Zagazig, Egypt
- *Correspondence: Kamal Awad
| | - Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Chair of Nephrology and Hypertension, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
- Maciej Banach
| |
Collapse
|
37
|
Wickramasinghe NM, Sachs D, Shewale B, Gonzalez DM, Dhanan-Krishnan P, Torre D, LaMarca E, Raimo S, Dariolli R, Serasinghe MN, Mayourian J, Sebra R, Beaumont K, Iyengar S, French DL, Hansen A, Eschenhagen T, Chipuk JE, Sobie EA, Jacobs A, Akbarian S, Ischiropoulos H, Ma'ayan A, Houten SM, Costa K, Dubois NC. PPARdelta activation induces metabolic and contractile maturation of human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2022; 29:559-576.e7. [PMID: 35325615 PMCID: PMC11072853 DOI: 10.1016/j.stem.2022.02.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/30/2021] [Accepted: 02/24/2022] [Indexed: 02/09/2023]
Abstract
Pluripotent stem-cell-derived cardiomyocytes (PSC-CMs) provide an unprecedented opportunity to study human heart development and disease, but they are functionally and structurally immature. Here, we induce efficient human PSC-CM (hPSC-CM) maturation through metabolic-pathway modulations. Specifically, we find that peroxisome-proliferator-associated receptor (PPAR) signaling regulates glycolysis and fatty acid oxidation (FAO) in an isoform-specific manner. While PPARalpha (PPARa) is the most active isoform in hPSC-CMs, PPARdelta (PPARd) activation efficiently upregulates the gene regulatory networks underlying FAO, increases mitochondrial and peroxisome content, enhances mitochondrial cristae formation, and augments FAO flux. PPARd activation further increases binucleation, enhances myofibril organization, and improves contractility. Transient lactate exposure, which is frequently used for hPSC-CM purification, induces an independent cardiac maturation program but, when combined with PPARd activation, still enhances oxidative metabolism. In summary, we investigate multiple metabolic modifications in hPSC-CMs and identify a role for PPARd signaling in inducing the metabolic switch from glycolysis to FAO in hPSC-CMs.
Collapse
Affiliation(s)
- Nadeera M Wickramasinghe
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Sachs
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David M Gonzalez
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Priyanka Dhanan-Krishnan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denis Torre
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth LaMarca
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Serena Raimo
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Madhavika N Serasinghe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua Mayourian
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristin Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Srinivas Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deborah L French
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Arne Hansen
- University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | | | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adam Jacobs
- Department of Obstetrics and Gynecology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Costa
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole C Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
38
|
Amat S, Dahlen CR, Swanson KC, Ward AK, Reynolds LP, Caton JS. Bovine Animal Model for Studying the Maternal Microbiome, in utero Microbial Colonization and Their Role in Offspring Development and Fetal Programming. Front Microbiol 2022; 13:854453. [PMID: 35283808 PMCID: PMC8916045 DOI: 10.3389/fmicb.2022.854453] [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: 01/14/2022] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Recent developments call for further research on the timing and mechanisms involved in the initial colonization of the fetal/infant gut by the maternal microbiome and its role in Developmental Origins of Health and Disease (DOHaD). Although progress has been made using primarily preterm infants, ethical and legal constraints hinder research progress in embryo/fetal-related research and understanding the developmental and mechanistic roles of the maternal microbiome in fetal microbial imprinting and its long-term role in early-life microbiome development. Rodent models have proven very good for studying the role of the maternal microbiome in fetal programming. However, some inherent limitations in these animal models make it challenging to study perinatal microbial colonization from a biomedical standpoint. In this review, we discuss the potential use of bovine animals as a biomedical model to study the maternal microbiome, in utero microbial colonization of the fetal gut, and their impact on offspring development and DOHaD.
Collapse
Affiliation(s)
- Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, United States
| | - Carl R Dahlen
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Kendall C Swanson
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Alison K Ward
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Lawrence P Reynolds
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Joel S Caton
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
39
|
Morris CC, Ref J, Acharya S, Johnson KJ, Squire S, Acharya T, Dennis T, Daugherty S, McArthur A, Chinyere IR, Koevary JW, Hare JM, Lancaster JJ, Goldman S, Avery R. Free-breathing gradient recalled echo-based CMR in a swine heart failure model. Sci Rep 2022; 12:3698. [PMID: 35260607 PMCID: PMC8904633 DOI: 10.1038/s41598-022-07611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 02/10/2022] [Indexed: 11/09/2022] Open
Abstract
In swine models, there are well-established protocols for creating a closed-chest myocardial infarction (MI) as well as protocols for characterization of cardiac function with cardiac magnetic resonance (CMR). This methods manuscript outlines a novel technique in CMR data acquisition utilizing smart-signal gradient recalled echo (GRE)-based array sequences in a free-breathing swine heart failure model allowing for both high spatial and temporal resolution imaging. Nine male Yucatan mini swine weighing 48.7 ± 1.6 kg at 58.2 ± 3.1 weeks old underwent the outlined imaging protocol before and 1-month after undergoing closed chest left anterior descending coronary artery (LAD) occlusion/reperfusion. The left ventricular ejection fraction (LVEF) at baseline was 59.3 ± 2.4% and decreased to 48.1 ± 3.7% 1-month post MI (P = 0.029). The average end-diastolic volume (EDV) at baseline was 55.2 ± 1.7 ml and increased to 74.2 ± 4.2 ml at 1-month post MI (P = 0.001). The resulting images from this novel technique and post-imaging analysis are presented and discussed. In a Yucatan swine model of heart failure via closed chest left anterior descending coronary artery (LAD) occlusion/reperfusion, we found that CMR with GRE-based array sequences produced clinical-grade images with high spatial and temporal resolution in the free-breathing setting.
Collapse
Affiliation(s)
- Craig C Morris
- Department of Medicine, Oregon Health and Sciences University, Portland, OR, USA
| | - Jacob Ref
- MD Program, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Satya Acharya
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Kevin J Johnson
- Magnetic Resonance Research Facility, University of Arizona, Tucson, AZ, USA
| | - Scott Squire
- Magnetic Resonance Research Facility, University of Arizona, Tucson, AZ, USA
| | | | - Tyler Dennis
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | | | - Alice McArthur
- Sarver Heart Center, University of Arizona, Tucson, AZ, USA
| | - Ikeotunye Royal Chinyere
- Sarver Heart Center, University of Arizona, Tucson, AZ, USA.,MD-PhD Program, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Jen Watson Koevary
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA
| | - Joshua M Hare
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Steven Goldman
- Sarver Heart Center, University of Arizona, Tucson, AZ, USA
| | - Ryan Avery
- Department of Radiology, Northwestern University, 676 N Saint Clair, Suite 800, Chicago, IL, 60611, USA.
| |
Collapse
|
40
|
Iborra-Egea O, Martínez-Falguera D, Roura S, Bayes-Genis A, Raya Á, Gálvez-Montón C. Porcine iPSC Generation: Testing Different Protocols to a Successful Application. Methods Mol Biol 2022; 2454:61-81. [PMID: 34845658 DOI: 10.1007/7651_2021_446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Stem cell therapy has an unparalleled potential to treat blood cancers, cardiovascular diseases and neurodegenerative conditions, among others. However, stem cell therapeutics must overcome multiple requirements before reaching clinical trials, including large animal safety and efficacy studies. In cardiovascular diseases swine models are the most widely adopted due to its great translational potential to humans. In this chapter, we will describe several protocols to induce iPSC dedifferentiation in swine fibroblasts, as well as conditioning treatments that may help in the reprogramming process.
Collapse
Affiliation(s)
- Oriol Iborra-Egea
- MyoCare Lab, ICREC Research Program, Germans Trias i Pujol Health Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Daina Martínez-Falguera
- MyoCare Lab, ICREC Research Program, Germans Trias i Pujol Health Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Santiago Roura
- MyoCare Lab, ICREC Research Program, Germans Trias i Pujol Health Research Institute (IGTP), Badalona, Barcelona, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayes-Genis
- MyoCare Lab, ICREC Research Program, Germans Trias i Pujol Health Research Institute (IGTP), Badalona, Barcelona, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- iCor Institute, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Ángel Raya
- Regenerative Medicine Program, Bellvitge Biomedical Research Institute (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), Hospital Duran i Reynals, Hospitalet de Llobregat, Barcelona, Spain
- Centre for Networked Biomedical Research on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Carolina Gálvez-Montón
- MyoCare Lab, ICREC Research Program, Germans Trias i Pujol Health Research Institute (IGTP), Badalona, Barcelona, Spain.
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
41
|
Martínez-Falguera D, Iborra-Egea O, Gálvez-Montón C. iPSC Therapy for Myocardial Infarction in Large Animal Models: Land of Hope and Dreams. Biomedicines 2021; 9:1836. [PMID: 34944652 PMCID: PMC8698445 DOI: 10.3390/biomedicines9121836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is the main driver of heart failure due to ischemia and subsequent cell death, and cell-based strategies have emerged as promising therapeutic methods to replace dead tissue in cardiovascular diseases. Research in this field has been dramatically advanced by the development of laboratory-induced pluripotent stem cells (iPSCs) that harbor the capability to become any cell type. Like other experimental strategies, stem cell therapy must meet multiple requirements before reaching the clinical trial phase, and in vivo models are indispensable for ensuring the safety of such novel therapies. Specifically, translational studies in large animal models are necessary to fully evaluate the therapeutic potential of this approach; to empirically determine the optimal combination of cell types, supplementary factors, and delivery methods to maximize efficacy; and to stringently assess safety. In the present review, we summarize the main strategies employed to generate iPSCs and differentiate them into cardiomyocytes in large animal species; the most critical differences between using small versus large animal models for cardiovascular studies; and the strategies that have been pursued regarding implanted cells' stage of differentiation, origin, and technical application.
Collapse
Affiliation(s)
- Daina Martínez-Falguera
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain;
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
42
|
Nutrition and Metabolism: Foundations for Animal Growth, Development, Reproduction, and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:1-24. [PMID: 34807434 DOI: 10.1007/978-3-030-85686-1_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Consumption of high-quality animal protein plays an important role in improving human nutrition, growth, development, and health. With an exponential growth of the global population, demands for animal-sourced protein are expected to increase by 60% between 2021 and 2050. In addition to the production of food protein and fiber (wool), animals are useful models for biomedical research to prevent and treat human diseases and serve as bioreactors to produce therapeutic proteins. For a high efficiency to transform low-quality feedstuffs and forages into high-quality protein and highly bioavailable essential minerals in diets of humans, farm animals have dietary requirements for energy, amino acids, lipids, carbohydrates, minerals, vitamins, and water in their life cycles. All nutrients interact with each other to influence the growth, development, and health of mammals, birds, fish, and crustaceans, and adequate nutrition is crucial for preventing and treating their metabolic disorders (including metabolic diseases) and infectious diseases. At the organ level, the small intestine is not only the terminal site for nutrient digestion and absorption, but also intimately interacts with a diverse community of intestinal antigens and bacteria to influence gut and whole-body health. Understanding the species and metabolism of intestinal microbes, as well as their interactions with the intestinal immune systems and the host intestinal epithelium can help to mitigate antimicrobial resistance and develop prebiotic and probiotic alternatives to in-feed antibiotics in animal production. As abundant sources of amino acids, bioactive peptides, energy, and highly bioavailable minerals and vitamins, animal by-product feedstuffs are effective for improving the growth, development, health, feed efficiency, and survival of livestock and poultry, as well as companion and aquatic animals. The new knowledge covered in this and related volumes of Adv Exp Med Biol is essential to ensure sufficient provision of animal protein for humans, while helping reduce greenhouse gas emissions, minimize the urinary and fecal excretion of nitrogenous and other wastes to the environment, and sustain animal agriculture (including aquaculture).
Collapse
|
43
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|
44
|
Kaldunski ML, Smith JR, Hayman GT, Brodie K, De Pons JL, Demos WM, Gibson AC, Hill ML, Hoffman MJ, Lamers L, Laulederkind SJF, Nalabolu HS, Thorat K, Thota J, Tutaj M, Tutaj MA, Vedi M, Wang SJ, Zacher S, Dwinell MR, Kwitek AE. The Rat Genome Database (RGD) facilitates genomic and phenotypic data integration across multiple species for biomedical research. Mamm Genome 2021; 33:66-80. [PMID: 34741192 PMCID: PMC8570235 DOI: 10.1007/s00335-021-09932-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/21/2021] [Indexed: 01/21/2023]
Abstract
Model organism research is essential for discovering the mechanisms of human diseases by defining biologically meaningful gene to disease relationships. The Rat Genome Database (RGD, ( https://rgd.mcw.edu )) is a cross-species knowledgebase and the premier online resource for rat genetic and physiologic data. This rich resource is enhanced by the inclusion and integration of comparative data for human and mouse, as well as other human disease models including chinchilla, dog, bonobo, pig, 13-lined ground squirrel, green monkey, and naked mole-rat. Functional information has been added to records via the assignment of annotations based on sequence similarity to human, rat, and mouse genes. RGD has also imported well-supported cross-species data from external resources. To enable use of these data, RGD has developed a robust infrastructure of standardized ontologies, data formats, and disease- and species-centric portals, complemented with a suite of innovative tools for discovery and analysis. Using examples of single-gene and polygenic human diseases, we illustrate how data from multiple species can help to identify or confirm a gene as involved in a disease and to identify model organisms that can be studied to understand the pathophysiology of a gene or pathway. The ultimate aim of this report is to demonstrate the utility of RGD not only as the core resource for the rat research community but also as a source of bioinformatic tools to support a wider audience, empowering the search for appropriate models for human afflictions.
Collapse
Affiliation(s)
- M L Kaldunski
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - J R Smith
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - G T Hayman
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - K Brodie
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - J L De Pons
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - W M Demos
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - A C Gibson
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M L Hill
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M J Hoffman
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - L Lamers
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - S J F Laulederkind
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - H S Nalabolu
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - K Thorat
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - J Thota
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Tutaj
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M A Tutaj
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Vedi
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - S J Wang
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
| | - S Zacher
- Information Services, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M R Dwinell
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - A E Kwitek
- Department of Biomedical Engineering, The Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
45
|
Genome editing in large animal models. Mol Ther 2021; 29:3140-3152. [PMID: 34601132 DOI: 10.1016/j.ymthe.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022] Open
Abstract
Although genome editing technologies have the potential to revolutionize the way we treat human diseases, barriers to successful clinical implementation remain. Increasingly, preclinical large animal models are being used to overcome these barriers. In particular, the immunogenicity and long-term safety of novel gene editing therapeutics must be evaluated rigorously. However, short-lived small animal models, such as mice and rats, cannot address secondary pathologies that may arise years after a gene editing treatment. Likewise, immunodeficient mouse models by definition lack the ability to quantify the host immune response to a novel transgene or gene-edited locus. Large animal models, including dogs, pigs, and non-human primates (NHPs), bear greater resemblance to human anatomy, immunology, and lifespan and can be studied over longer timescales with clinical dosing regimens that are more relevant to humans. These models allow for larger scale and repeated blood and tissue sampling, enabling greater depth of study and focus on rare cellular subsets. Here, we review current progress in the development and evaluation of novel genome editing therapies in large animal models, focusing on applications in human immunodeficiency virus 1 (HIV-1) infection, cancer, and genetic diseases including hemoglobinopathies, Duchenne muscular dystrophy (DMD), hypercholesterolemia, and inherited retinal diseases.
Collapse
|
46
|
Morel S, Schilling S, Diagbouga MR, Delucchi M, Bochaton-Piallat ML, Lemeille S, Hirsch S, Kwak BR. Effects of Low and High Aneurysmal Wall Shear Stress on Endothelial Cell Behavior: Differences and Similarities. Front Physiol 2021; 12:727338. [PMID: 34721060 PMCID: PMC8551710 DOI: 10.3389/fphys.2021.727338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Intracranial aneurysms (IAs) result from abnormal enlargement of the arterial lumen. IAs are mostly quiescent and asymptomatic, but their rupture leads to severe brain damage or death. As the evolution of IAs is hard to predict and intricates medical decision, it is essential to improve our understanding of their pathophysiology. Wall shear stress (WSS) is proposed to influence IA growth and rupture. In this study, we investigated the effects of low and supra-high aneurysmal WSS on endothelial cells (ECs). Methods: Porcine arterial ECs were exposed for 48 h to defined levels of shear stress (2, 30, or 80 dyne/cm2) using an Ibidi flow apparatus. Immunostaining for CD31 or γ-cytoplasmic actin was performed to outline cell borders or to determine cell architecture. Geometry measurements (cell orientation, area, circularity and aspect ratio) were performed on confocal microscopy images. mRNA was extracted for RNAseq analysis. Results: ECs exposed to low or supra-high aneurysmal WSS were more circular and had a lower aspect ratio than cells exposed to physiological flow. Furthermore, they lost the alignment in the direction of flow observed under physiological conditions. The effects of low WSS on differential gene expression were stronger than those of supra-high WSS. Gene set enrichment analysis highlighted that extracellular matrix proteins, cytoskeletal proteins and more particularly the actin protein family were among the protein classes the most affected by shear stress. Interestingly, most genes showed an opposite regulation under both types of aneurysmal WSS. Immunostainings for γ-cytoplasmic actin suggested a different organization of this cytoskeletal protein between ECs exposed to physiological and both types of aneurysmal WSS. Conclusion: Under both aneurysmal low and supra-high WSS the typical arterial EC morphology molds to a more spherical shape. Whereas low WSS down-regulates the expression of cytoskeletal-related proteins and up-regulates extracellular matrix proteins, supra-high WSS induces opposite changes in gene expression of these protein classes. The differential regulation in EC gene expression observed under various WSS translate into a different organization of the ECs’ architecture. This adaptation of ECs to different aneurysmal WSS conditions may affect vascular remodeling in IAs.
Collapse
Affiliation(s)
- Sandrine Morel
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Neurosurgery Division, Department of Clinical Neurosciences, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Sabine Schilling
- Institute of Applied Simulation, Zurich University of Applied Sciences, Wädenswil, Switzerland.,Institute of Tourism and Mobility, Lucerne School of Business, Lucerne University of Applied Sciences and Arts, Lucerne, Switzerland
| | - Mannekomba R Diagbouga
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Matteo Delucchi
- Institute of Applied Simulation, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | | | - Sylvain Lemeille
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sven Hirsch
- Institute of Applied Simulation, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
47
|
Jiang Y, Zou Q, Liu B, Li S, Wang Y, Liu T, Ding X. Atlas of Prenatal Hair Follicle Morphogenesis Using the Pig as a Model System. Front Cell Dev Biol 2021; 9:721979. [PMID: 34692680 PMCID: PMC8529045 DOI: 10.3389/fcell.2021.721979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/13/2021] [Indexed: 01/15/2023] Open
Abstract
The pig is an increasingly popular biomedical model, but only a few in depth data exist on its studies in hair follicle (HF) morphogenesis and development. Hence, the objective of this study was to identify the suitability of the pig as an animal model for human hair research. We performed a classification of pig HF morphogenesis stages and hair types. All four different hair types sampled from 17 different body parts in pig were similar to those of human. The Guard_2 sub-type was more similar to type II human scalp hair while Guard_1, Awl, Auchene, and Zigzag were similar to type I scalp hair. Based on morphological observation and marker gene expression of HF at 11 different embryonic days and six postnatal days, we classified pig HF morphogenesis development from E41 to P45 into three main periods - induction (E37-E41), organogenesis (E41-E85), and cytodifferentiation (>E85). Furthermore, we demonstrated that human and pig share high similarities in HF morphogenesis occurrence time (early/mid gestational) and marker gene expression patterns. Our findings will facilitate the study of human follicle morphogenesis and research on complex hair diseases and offer researchers a suitable model for human hair research.
Collapse
Affiliation(s)
- Yao Jiang
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Quan Zou
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Bo Liu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shujuan Li
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi Wang
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tianlong Liu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangdong Ding
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
48
|
Dittfeld C, Winkelkotte M, Behrens S, Schmieder F, Jannasch A, Matschke K, Sonntag F, Tugtekin SM. Establishment of a resazurin-based aortic valve tissue viability assay for dynamic culture in a microphysiological system. Clin Hemorheol Microcirc 2021; 79:167-178. [PMID: 34487029 DOI: 10.3233/ch-219112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/AIM Tissue pathogenesis of aortic valve (AV) stenosis is research focus in cardiac surgery. Model limitations of conventional 2D culture of human or porcine valvular interstitial/endothelial cells (VIC/VECs) isolated from aortic valve tissues but also limited ability of (small) animal models to reflect human (patho)physiological situation in AV position raise the need to establish an in vitro setup using AV tissues. Resulting aim is to approximate (patho)physiological conditions in a dynamic pulsatile Microphysiological System (MPS) to culture human and porcine AV tissue with preservation of tissue viability but also defined ECM composition. MATERIALS/METHODS A tissue incubation chamber (TIC) was designed to implement human or porcine tissues (3×5 mm2) in a dynamic pulsatile culture in conventional cell culture ambience in a MPS. Cell viability assays based on lactate dehydrogenase (LDH)-release or resazurin-conversion were tested for applicability in the system and applied for a culture period of 14 days with interval evaluation of tissue viability on every other day. Resazurin-assay setup was compared in static vs. dynamic culture using varying substance saturation settings (50-300μM), incubation times and tissue masses and was consequently adapted. RESULTS Sterile dynamic culture of human and porcine AV tissue segments was established at a pulsatile flow rate range of 0.9-13.4μl/s. Implementation of tissues was realized by stitching the material in a thermoplastic polyurethane (TPU)-ring and insertion in the TIC-MPS-system. Culture volume of 2 ml caused LDH dilution not detectable in standard membrane integrity assay setup. Therefore, detection of resazurin-conversion of viable tissue was investigated. Optimal incubation time for viability conversion was determined at two hours at a saturated concentration of 300μM resazurin. Measurement in static conditions was shown to offer comparable results as dynamic condition but allowing optimal handling and TIC sterilization protocols for long term culture. Preliminary results revealed favourable porcine AV tissue viability over a 14 day period confirmed via resazurin-assay comparing statically cultured tissue counterparts. CONCLUSIONS Human and porcine AV tissue can be dynamically cultured in a TIC-MPS with monitoring of tissue viability using an adapted resazurin-assay setup. Preliminary results reveal advantageous viability of porcine AV tissues after dynamic TIC-MPS culture compared to static control.
Collapse
Affiliation(s)
- C Dittfeld
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Dresden,Germany
| | - M Winkelkotte
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Dresden,Germany
| | - S Behrens
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - F Schmieder
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - A Jannasch
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Dresden,Germany
| | - K Matschke
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Dresden,Germany
| | - F Sonntag
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - S M Tugtekin
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Dresden,Germany
| |
Collapse
|
49
|
Jang DH, Piel S, Greenwood JC, Kelly M, Mazandi VM, Ranganathan A, Lin Y, Starr J, Hallowell T, Shofer FS, Baker WB, Lafontant A, Andersen K, Ehinger JK, Kilbaugh TJ. Alterations in cerebral and cardiac mitochondrial function in a porcine model of acute carbon monoxide poisoning. Clin Toxicol (Phila) 2021; 59:801-809. [PMID: 33529085 PMCID: PMC8326298 DOI: 10.1080/15563650.2020.1870691] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 12/25/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVES The purpose of this study is the development of a porcine model of carbon monoxide (CO) poisoning to investigate alterations in brain and heart mitochondrial function. DESIGN Two group large animal model of CO poisoning. SETTING Laboratory. SUBJECTS Ten swine were divided into two groups: Control (n = 4) and CO (n = 6). INTERVENTIONS Administration of a low dose of CO at 200 ppm to the CO group over 90 min followed by 30 min of re-oxygenation at room air. The Control group received room air for 120 min. MEASUREMENTS Non-invasive optical monitoring was used to measure cerebral blood flow and oxygenation. Cerebral microdialysis was performed to obtain semi real time measurements of cerebral metabolic status. At the end of the exposure, both fresh brain (cortical and hippocampal tissue) and heart (apical tissue) were immediately harvested to measure mitochondrial respiration and reactive oxygen species (ROS) generation and blood was collected to assess plasma cytokine concentrations. MAIN RESULTS Animals in the CO group showed significantly decreased Complex IV-linked mitochondrial respiration in hippocampal and apical heart tissue but not cortical tissue. There also was a significant increase in mitochondrial ROS generation across all measured tissue types. The CO group showed a significantly higher cerebral lactate-to-pyruvate ratio. Both IL-8 and TNFα were significantly increased in the CO group compared with the Control group obtained from plasma. While not significant there was a trend to an increase in optically measured cerebral blood flow and hemoglobin concentration in the CO group. CONCLUSIONS Low-dose CO poisoning is associated with early mitochondrial disruption prior to an observable phenotype highlighting the important role of mitochondrial function in the pathology of CO poisoning. This may represent an important intervenable pathway for therapy and intervention.
Collapse
Affiliation(s)
- David H. Jang
- Department of Emergency Medicine, Division of Medical Toxicology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sarah Piel
- Resuscitation Science Center, Philadelphia, PA, USA
| | - John C. Greenwood
- Department of Anesthesiology and Critical Care Medicine, Department of Emergency Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Matthew Kelly
- Department of Emergency Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Yuxi Lin
- Resuscitation Science Center, Philadelphia, PA, USA
| | | | | | - Frances S. Shofer
- Department of Emergency Medicine, Division of Medical Toxicology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Wesley B. Baker
- Department of Pediatric Neurology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Alec Lafontant
- Department of Pediatric Neurology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Kristen Andersen
- Department of Pediatric Neurology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Johannes K. Ehinger
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Skåne University Hospital, Lund University, Malmo, Sweden
| | | |
Collapse
|
50
|
Ayuso SA, Shipp RC, Aladegbami BG, Farquharson D, Lawson D, Bassett R. AbsorbaSeal™ 5.6.7F vascular closure device: A good laboratory practice chronic study evaluating safety and efficacy in a healthy porcine model. Vascular 2021; 30:934-942. [PMID: 34459306 DOI: 10.1177/17085381211037883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Vascular closure devices (VCDs) are widely used for arteriotomy closure after percutaneous catheter-based procedures. In comparison to manual compression, VCDs have been associated with shorter time to hemostasis, shorter time to ambulation, and also decreased length of stay. Complexity of deployment, lack of immediate hemostasis, and residual deformity of the arterial wall remain as limitations of current VCDs. The aim of this study was to investigate the AbsorbaSeal™ 5.6.7F vascular closure device, a novel, completely bioabsorbable, intuitive, and easy to use VCD which uses a compressive, "sandwich"-type design comprising a low profile intravascular distal seal and gasket and an extravascular floating foot and proximal seal, in an open infrarenal aortic swine model. METHODS Eight fully heparinized swine at a good laboratory practices facility underwent AbsorbaSeal™ 5.6.7F VCD closure of three 6F arteriotomies each in the proximal, mid, and distal infrarenal aorta. Two swine underwent harvest at each of four time cohorts: 30, 60, 90, and 120 days. Acute and chronic procedural safety and efficacy, as well as target site vascular remodeling over time, were the primary outcomes evaluated. Secondary outcome measures included local and systemic inflammatory responses, end-organ tissue analysis, and device-related complications through the follow-up periods. Histopathological evaluation was performed by a blinded pathologist. Standard statistical methods were used. RESULTS In deployment of 24 AbsorbaSeal™ 5.6.7F VCDs, there were no device-related complications or mortalities. All deployments resulted in rapid arteriotomy seal (100% deployment success), with a mean time to hemostasis (cessation of arterial flow) of 21.5 s (median: 6.5 s) across a mean activated clotting time (ACT) of 356 s. Twenty of the 24 implant sites (83%) attained complete hemostasis within 20 s. Immediate post-implant and pre-termination angiographies at all time points were performed of all swine which demonstrated normal aortic appearance and tissue structure and normal downstream vascular beds. At 30 days, each implant's intravascular distal seal and gasket were removed from the circulation and completely covered with a smooth neointimal layer. Minimal inflammation and no intimal or luminal thrombus were observed at any site at every time point. CONCLUSIONS AbsorbaSeal™ 5.6.7F is a safe, effective, and secure VCD that demonstrates rapid hemostasis in a fully heparinized open aortic porcine model. Removal from circulation and complete coverage of the intravascular distal seal and gasket with neointima occurred within 30 days post-implant. Natural transmural vessel healing from the arteriotomy itself with minimal inflammation was noted for each implant at every time point.
Collapse
Affiliation(s)
- Sullivan A Ayuso
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, 22442Carolinas Medical Center, Charlotte, NC, USA
| | - R Caroline Shipp
- 171799The University of Tennessee at Chattanooga College of Arts and Sciences, Chattanooga, TN, USA
| | - Bola G Aladegbami
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, 22442Carolinas Medical Center, Charlotte, NC, USA
| | - Delton Farquharson
- General and Vascular Surgery, 58810Princess Margaret Hospital, Nassau, Bahamas
| | | | | |
Collapse
|