1
|
Jeung S, An JH, Kim SS, Youn HY. Safety and efficacy of canine gonadal tissue-derived mesenchymal stem cells for early myxomatous mitral valve disease. Front Vet Sci 2024; 11:1404607. [PMID: 39415950 PMCID: PMC11480051 DOI: 10.3389/fvets.2024.1404607] [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: 03/21/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction This study explored the potential efficacy and safety of therapy with mesenchymal stem cells (MSC) derived from gonadal tissue to address the early stage of myxomatous mitral valve disease (MMVD), the predominant cardiac condition in dogs. Methods Sixteen dogs diagnosed with MMVD B1 were enrolled in this trial and assigned to either a control group (control group, n = 10) or a group that received MSC derived from gonadal tissue (treatment group, n = 6). In the treatment group, allogeneic MSC derived from gonadal tissue (1 × 106 cells/kg) were intravenously administered at monthly intervals for five or more sessions. Data were compared at baseline and at the endpoint 1-year intervals. The efficacy was assessed using echocardiography, thoracic radiography, NT-proBNP, and the duration from B1 diagnosis to B2 transition to evaluate its effect on MMVD stage progression. Safety was evaluated through physical examinations, blood tests, imaging studies, and monitoring of adverse events. Results After 1 year of observation, the control group exhibited deteriorating echocardiographic parameters, whereas the treatment group displayed no substantial differences between baseline and endpoint measurements. Notably, a statistically significant disparity was noted in the left atrial diameter (p < 0.05) and E-wave velocity (p < 0.05) between the two groups, indicating a favorable impact of MSC derived from the gonadal tissue on left atrial pressure. Additionally, in contrast to the control group, the treatment group demonstrated delayed progression to MMVD stage B2, enabling them to prolong their disease duration without requiring cardiac medication (p = 0.038). In quality of life (QoL) metrics following MSC treatment, appetite showed a statistically significant improvement, increasing from 4 to 4.83 (p < 0.05). Discussion Treatment with gonadal tissue-derived MSCs significantly delayed MMVD stage progression, highlighting the broad potential of MSC derived from gonadal tissue for treating complex veterinary conditions.
Collapse
Affiliation(s)
- Soyoung Jeung
- VIP Animal Medical Center, Seoul, Republic of Korea
- Laboratory of Veterinary Internal Medicine, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ju-Hyun An
- Laboratory of Veterinary Emergency and Critical Care, Department of Veterinary Clinical Science, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Sung-Soo Kim
- VIP Animal Medical Center, Seoul, Republic of Korea
- Laboratory of Veterinary Internal Medicine, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Williams ZJ, Pezzanite LM, Chow L, Rockow M, Dow SW. Evaluation of stem-cell therapies in companion animal disease models: a concise review (2015-2023). Stem Cells 2024; 42:677-705. [PMID: 38795363 DOI: 10.1093/stmcls/sxae034] [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: 11/26/2023] [Accepted: 01/25/2024] [Indexed: 05/27/2024]
Abstract
Companion animals in veterinary medicine develop multiple naturally occurring diseases analogous to human conditions. We previously reported a comprehensive review on the feasibility, safety, and biologic activity of using novel stem cell therapies to treat a variety of inflammatory conditions in dogs and cats (2008-2015) [Hoffman AM, Dow SW. Concise review: stem cell trials using companion animal disease models. Stem Cells. 2016;34(7):1709-1729. https://doi.org/10.1002/stem.2377]. The purpose of this review is to provide an updated summary of current studies in companion animal disease models that have evaluated stem cell therapeutics that are relevant to human disease. Here we have reviewed the literature from 2015 to 2023 for publications on stem cell therapies that have been evaluated in companion animals, including dogs, cats, and horses. The review excluded case reports or studies performed in experimentally induced models of disease, studies involving cancer, or studies in purpose-bred laboratory species such as rodents. We identified 45 manuscripts meeting these criteria, an increase from 19 that were described in the previous review [Hoffman AM, Dow SW. Concise review: stem cell trials using companion animal disease models. Stem Cells. 2016;34(7):1709-1729. https://doi.org/10.1002/stem.2377]. The majority of studies were performed in dogs (n = 28), with additional studies in horses (n = 9) and cats (n = 8). Disease models included those related to musculoskeletal disease (osteoarthritis and tendon/ligament injury), neurologic disease (canine cognitive dysfunction, intervertebral disc disease, spinal cord injury) gingival/dental disease (gingivostomatitis), dermatologic disease (atopic dermatitis), chronic multi-drug resistant infections, ophthalmic disease (keratoconjunctivitis sicca, eosinophilic keratitis, immune-mediated keratitis), cardiopulmonary disease (asthma, degenerative valve disease, dilated cardiomyopathy), gastrointestinal disease (inflammatory bowel disease, chronic enteropathy), and renal disease (chronic kidney disease). The majority of studies reported beneficial responses to stem cell treatment, with the exception of those related to more chronic processes such as spinal cord injury and chronic kidney disease. However, it should also be noted that 22 studies were open-label, baseline-controlled trials and only 12 studies were randomized and controlled, making overall study interpretation difficult. As noted in the previous review, improved regulatory oversight and consistency in manufacturing of stem cell therapies are needed. Enhanced understanding of the temporal course of disease processes using advanced-omics approaches may further inform mechanisms of action and help define appropriate timing of interventions. Future directions of stem-cell-based therapies could include use of stem-cell-derived extracellular vesicles, or cell conditioning approaches to direct cells to specific pathways that are tailored to individual disease processes and stages of illness.
Collapse
Affiliation(s)
- Zoë J Williams
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Meagan Rockow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Steven W Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
3
|
El-Husseiny HM, Mady EA, Helal MAY, Tanaka R. The Pivotal Role of Stem Cells in Veterinary Regenerative Medicine and Tissue Engineering. Vet Sci 2022; 9:648. [PMID: 36423096 PMCID: PMC9698002 DOI: 10.3390/vetsci9110648] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 07/30/2023] Open
Abstract
The introduction of new regenerative therapeutic modalities in the veterinary practice has recently picked up a lot of interest. Stem cells are undifferentiated cells with a high capacity to self-renew and develop into tissue cells with specific roles. Hence, they are an effective therapeutic option to ameliorate the ability of the body to repair and engineer damaged tissues. Currently, based on their facile isolation and culture procedures and the absence of ethical concerns with their use, mesenchymal stem cells (MSCs) are the most promising stem cell type for therapeutic applications. They are becoming more and more well-known in veterinary medicine because of their exceptional immunomodulatory capabilities. However, their implementation on the clinical scale is still challenging. These limitations to their use in diverse affections in different animals drive the advancement of these therapies. In the present article, we discuss the ability of MSCs as a potent therapeutic modality for the engineering of different animals' tissues including the heart, skin, digestive system (mouth, teeth, gastrointestinal tract, and liver), musculoskeletal system (tendons, ligaments, joints, muscles, and nerves), kidneys, respiratory system, and eyes based on the existing knowledge. Moreover, we highlighted the promises of the implementation of MSCs in clinical use in veterinary practice.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Mahmoud A. Y. Helal
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Animal Medicine, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
| |
Collapse
|
4
|
Liu J, Ren Z, Yang L, Zhu L, li Y, Bie C, Liu H, Ji Y, Chen D, Zhu M, Kuang W. The NSUN5-FTH1/FTL pathway mediates ferroptosis in bone marrow-derived mesenchymal stem cells. Cell Death Dis 2022; 8:99. [PMID: 35249107 PMCID: PMC8898311 DOI: 10.1038/s41420-022-00902-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/16/2021] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
AbstractFerroptosis is a type of cell death induced by the iron-dependent accumulation of lipid hydroperoxides and reactive oxygen species (ROS) in cells. Inhibiting ferroptosis is important for improving the survival of transplanted bone marrow-derived mesenchymal stem cells (BMSCs). Although it is known that NOP2/Sun RNA methyltransferase 5 (NSUN5) post-transcriptionally regulates ferroptosis in BMSCs through RNA methylation, the precise mechanisms underlying these effects have not been reported. In this study, we demonstrate that NSUN5 is downregulated in erastin-induced ferroptosis in BMSCs. Ferroptosis was inhibited by the overexpression of NSUN5 or ferritin heavy chain/light-chain (FTH1/FTL) and was enhanced by NSUN5 knockdown. RNA immunoprecipitation experiments revealed that NSUN5 binds to FTH1/FTL, while NSUN5 depletion reduced the levels of 5-methylcytosine in FTH1/FTL RNA and increased intracellular iron concentrations, resulting in the downregulation of glutathione peroxidase 4 (GPX4) and the accumulation of ROS and lipid peroxidation products. Co-immunoprecipitation experiments demonstrated that the recognition of FTH1 and FTL by NSUN5 is dependent on the recruitment of tumor necrosis factor receptor-associated protein 1 (TRAP1). These results suggested that the NSUN5-FTH1/FTL pathway mediates ferroptosis in BMSCs and that the therapeutic targeting of components of this pathway may promote resistance to ferroptosis and improve the survival of transplanted BMSCs.
Collapse
|
5
|
Zhu D, Cheng K. Cardiac Cell Therapy for Heart Repair: Should the Cells Be Left Out? Cells 2021; 10:641. [PMID: 33805763 PMCID: PMC7999733 DOI: 10.3390/cells10030641] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is still the leading cause of death worldwide. Coronary artery occlusion, or myocardial infarction (MI) causes massive loss of cardiomyocytes. The ischemia area is eventually replaced by a fibrotic scar. From the mechanical dysfunctions of the scar in electronic transduction, contraction and compliance, pathological cardiac dilation and heart failure develops. Once end-stage heart failure occurs, the only option is to perform heart transplantation. The sequential changes are termed cardiac remodeling, and are due to the lack of endogenous regenerative actions in the adult human heart. Regenerative medicine and biomedical engineering strategies have been pursued to repair the damaged heart and to restore normal cardiac function. Such strategies include both cellular and acellular products, in combination with biomaterials. In addition, substantial progress has been made to elucidate the molecular and cellular mechanisms underlying heart repair and regeneration. In this review, we summarize and discuss current therapeutic approaches for cardiac repair and provide a perspective on novel strategies that holding potential opportunities for future research and clinical translation.
Collapse
Affiliation(s)
- Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
6
|
Kang MH, Park HM. Challenges of stem cell therapies in companion animal practice. J Vet Sci 2020; 21:e42. [PMID: 32476316 PMCID: PMC7263915 DOI: 10.4142/jvs.2020.21.e42] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine using stem cells from various sources are emerging treatment modality in several refractory diseases in veterinary medicine. It is well-known that stem cells can differentiate into specific cell types, self-renew, and regenerate. In addition, the unique immunomodulatory effects of stem cells have made stem cell transplantation a promising option for treating a wide range of disease and injuries. Recently, the medical demands for companion animals have been rapidly increasing, and certain disease conditions require alternative treatment options. In this review, we focused on stem cell application research in companion animals including experimental models, case reports and clinical trials in dogs and cats. The clinical studies and therapeutic protocols were categorized, evaluated and summarized according to the organ systems involved. The results indicate that evidence for the effectiveness of cell-based treatment in specific diseases or organ systems is not yet conclusive. Nonetheless, stem cell therapy may be a realistic treatment option in the near future, therefore, considerable efforts are needed to find optimized cell sources, cell numbers and delivery methods in order to standardize treatment methods and evaluation processes.
Collapse
Affiliation(s)
- Min Hee Kang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Hee Myung Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
7
|
Species differences in cardiovascular physiology that affect pharmacology and toxicology. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
8
|
Ribitsch I, Baptista PM, Lange-Consiglio A, Melotti L, Patruno M, Jenner F, Schnabl-Feichter E, Dutton LC, Connolly DJ, van Steenbeek FG, Dudhia J, Penning LC. Large Animal Models in Regenerative Medicine and Tissue Engineering: To Do or Not to Do. Front Bioeng Biotechnol 2020; 8:972. [PMID: 32903631 PMCID: PMC7438731 DOI: 10.3389/fbioe.2020.00972] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Rapid developments in Regenerative Medicine and Tissue Engineering has witnessed an increasing drive toward clinical translation of breakthrough technologies. However, the progression of promising preclinical data to achieve successful clinical market authorisation remains a bottleneck. One hurdle for progress to the clinic is the transition from small animal research to advanced preclinical studies in large animals to test safety and efficacy of products. Notwithstanding this, to draw meaningful and reliable conclusions from animal experiments it is critical that the species and disease model of choice is relevant to answer the research question as well as the clinical problem. Selecting the most appropriate animal model requires in-depth knowledge of specific species and breeds to ascertain the adequacy of the model and outcome measures that closely mirror the clinical situation. Traditional reductionist approaches in animal experiments, which often do not sufficiently reflect the studied disease, are still the norm and can result in a disconnect in outcomes observed between animal studies and clinical trials. To address these concerns a reconsideration in approach will be required. This should include a stepwise approach using in vitro and ex vivo experiments as well as in silico modeling to minimize the need for in vivo studies for screening and early development studies, followed by large animal models which more closely resemble human disease. Naturally occurring, or spontaneous diseases in large animals remain a largely untapped resource, and given the similarities in pathophysiology to humans they not only allow for studying new treatment strategies but also disease etiology and prevention. Naturally occurring disease models, particularly for longer lived large animal species, allow for studying disorders at an age when the disease is most prevalent. As these diseases are usually also a concern in the chosen veterinary species they would be beneficiaries of newly developed therapies. Improved awareness of the progress in animal models is mutually beneficial for animals, researchers, human and veterinary patients. In this overview we describe advantages and disadvantages of various animal models including domesticated and companion animals used in regenerative medicine and tissue engineering to provide an informed choice of disease-relevant animal models.
Collapse
Affiliation(s)
- Iris Ribitsch
- Veterm, Department for Companion Animals and Horses, University Equine Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Pedro M. Baptista
- Laboratory of Organ Bioengineering and Regenerative Medicine, Health Research Institute of Aragon (IIS Aragon), Zaragoza, Spain
| | - Anna Lange-Consiglio
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Luca Melotti
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Florien Jenner
- Veterm, Department for Companion Animals and Horses, University Equine Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva Schnabl-Feichter
- Clinical Unit of Small Animal Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Luke C. Dutton
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - David J. Connolly
- Clinical Unit of Small Animal Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jayesh Dudhia
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
9
|
Voga M, Adamic N, Vengust M, Majdic G. Stem Cells in Veterinary Medicine-Current State and Treatment Options. Front Vet Sci 2020; 7:278. [PMID: 32656249 PMCID: PMC7326035 DOI: 10.3389/fvets.2020.00278] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Regenerative medicine is a branch of medicine that develops methods to grow, repair, or replace damaged or diseased cells, organs or tissues. It has gained significant momentum in recent years. Stem cells are undifferentiated cells with the capability to self—renew and differentiate into tissue cells with specialized functions. Stem cell therapies are therefore used to overcome the body's inability to regenerate damaged tissues and metabolic processes after acute or chronic insult. The concept of stem cell therapy was first introduced in 1991 by Caplan, who proposed that massive differentiation of cells into the desired tissue could be achieved by isolation, cultivation, and expansion of stem cells in in vitro conditions. Among different stem cell types, mesenchymal stem cells (MSC) currently seem to be the most suitable for therapeutic purposes, based on their simple isolation and culturing techniques, and lack of ethical issues regarding their usage. Because of their remarkable immunomodulatory abilities, MSCs are increasingly gaining recognition in veterinary medicine. Developments are primarily driven by the limitations of current treatment options for various medical problems in different animal species. MSCs represent a possible therapeutic option for many animal diseases, such as orthopedic, orodental and digestive tract diseases, liver, renal, cardiac, respiratory, neuromuscular, dermal, olfactory, and reproductive system diseases. Although we are progressively gaining an understanding of MSC behavior and their mechanisms of action, some of the issues considering their use for therapy are yet to be resolved. The aim of this review is first to summarize the current knowledge and stress out major issues in stem cell based therapies in veterinary medicine and, secondly, to present results of clinical usage of stem cells in veterinary patients.
Collapse
Affiliation(s)
- Metka Voga
- Faculty of Veterinary Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Neza Adamic
- Faculty of Veterinary Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Modest Vengust
- Faculty of Veterinary Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
10
|
Müller-Ruch U, Skorska A, Lemcke H, Steinhoff G, David R. GLP: A requirement in cell therapies - perspectives for the cardiovascular field. Adv Drug Deliv Rev 2020; 165-166:96-104. [PMID: 32305352 DOI: 10.1016/j.addr.2020.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 02/08/2023]
Abstract
In biomedical research, enormous progress is being made and new candidates for putative medicinal products emerge. However, most published preclinical data are not conducted according to the standard Good Laboratory Practice (GLP). GLP is mandatory for preclinical analysis of Advanced Therapy Medicinal Products (ATMP) and thereby a prerequisite for planning and conduction of clinical trials. Not inconsiderable numbers of clinical trials are terminated earlier or fail - do inadequate testing strategies or missing specialized assays during the preclinical development contribute to this severe complex of problems? Unfortunately, there is also a lack of access to GLP testing results and OECD (Organisation for Economic Co-operation and Development) GLP guidelines are not yet adjusted to ATMP specialties. Ultimately, GLP offers possibilities to generate reliable and reproducible data. Therefore, this review elucidates different GLP aspects in drug development, speculates on reasons of putative low GLP acceptance in the scientific community and mentions solution proposals.
Collapse
|
11
|
Werner S, Wallukat G, Becker NP, Wenzel K, Müller J, Schimke I, Wess G. The aptamer BC 007 for treatment of dilated cardiomyopathy: evaluation in Doberman Pinschers of efficacy and outcomes. ESC Heart Fail 2020; 7:844-855. [PMID: 32212256 PMCID: PMC7261533 DOI: 10.1002/ehf2.12628] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/14/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Aims Aptamer BC 007, a 15‐mer single‐strand DNA oligonucleotide (5'‐GGTTGGTGTGGTTGG‐3'), was developed to neutralize functional autoantibodies that bind to the extracellular domains of G protein‐coupled receptors (GPCR‐AAB), leading to the modulation of receptor‐mediated signalling cascades that induce pathophysiological states. Among the GPCR‐AAB, there are those directed against the β1‐adrenergic receptor (β1‐AAB) that are highly present in patients with dilated cardiomyopathy (DCM) and are increasingly accepted as disease drivers. Using Doberman Pinschers (DP) with DCM, which possess similarities with human DCM among these β1‐AAB positivity for that the disease‐driving role in DP DCM was demonstrated, the safety of BC 007, efficacy for neutralizing β1‐AAB, and the DP's outcome were investigated. Methods and results Fourteen client‐owned β1‐AAB‐positive DP with electrocardiographically and echocardiographically indicated DCM were treated with BC 007. For controlling, two groups were created: 14 β1‐AAB‐positive DP with DCM not treated with BC 007 (Control 1) and 14 DP with DCM closely matched to the BC 007‐treated DP (Control 2), retrospectively selected from the institutional database of DP. After treatment, DP were monitored both echocardiographically, and for β1‐AAB, and survival curves were calculated. Based on clinical and laboratory examination, no adverse effects associated with BC 007 treatment were observed during the study. Forty‐eight hours after treatment, the DP's blood was free of β1‐AAB, which led to a reduction or stabilization of left ventricular end‐systolic volume (ESVI) during β1‐AAB free time in 10 of the treated DP. In one DP, where β1‐AAB returned after 3 months and ESVI worsened again, a second BC 007 treatment after 9 months again cleared the blood from β1‐AAB and improved the ESVI. Compared with the controls, DP treated with BC 007 showed a significantly longer survival time [572 days, interquartile range (IQR) 442–840 days] vs. Control group 1 (266 days, IQR 97–438 days; logrank: P = 0.009) and Control group 2 (229 days, IQR 174–319 days; logrank: P = 0.012). Conclusions Treatment with BC 007 for β1‐AAB neutralization was safe, resulted in a long‐lasting reduction of β1‐AAB combined with improved cardiac function and prolonged the survival of DP with DCM. Using a natural large animal model of DCM considered superior to small animal models of immunization‐induced cardiomyopathy, combined with a study design comparable with clinical trials, we believe that our results provide the basis for optimism that treatment with BC 007 might also be effective in human patients with DCM.
Collapse
Affiliation(s)
- Sabine Werner
- Department of Cardiology, Clinic of Small Animal Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Gerd Wallukat
- Department of Research & Development, Berlin Cures GmbH, Berlin, Germany
| | - Niels-Peter Becker
- Department of Research & Development, Berlin Cures GmbH, Berlin, Germany
| | - Katrin Wenzel
- Department of Research & Development, Berlin Cures GmbH, Berlin, Germany
| | - Johannes Müller
- Department of Research & Development, Berlin Cures GmbH, Berlin, Germany
| | - Ingolf Schimke
- Department of Research & Development, Berlin Cures GmbH, Berlin, Germany
| | - Gerhard Wess
- Department of Cardiology, Clinic of Small Animal Medicine, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
12
|
Wang S, Chen W, Ma L, Zou M, Dong W, Yang H, Sun L, Chen X, Duan J. Infant cardiosphere-derived cells exhibit non-durable heart protection in dilated cardiomyopathy rats. Cytotechnology 2019; 71:1043-1052. [PMID: 31583508 DOI: 10.1007/s10616-019-00328-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/25/2019] [Indexed: 12/18/2022] Open
Abstract
Stem cells provide a new strategy for the treatment of cardiac diseases; however, their effectiveness in dilated cardiomyopathy (DCM) has not been investigated. In this study, cardiosphere-derived cells (CDCs) were isolated from infants (≤ 24 months) and identified by the cell surface markers CD105, CD90, CD117 and CD45, which is consistent with a previous report, although increased CD34 expression was observed. The molecular expression profile of CDCs from infants was determined by RNA sequencing and compared with adult CDCs, showing that infant CDCs have almost completely altered gene expression patterns compared with adult CDCs. The upregulated genes in infant CDCs are mainly related to the biological processes of cell morphogenesis and differentiation. The molecular profile of infant CDCs was characterized by lower expression of inflammatory cytokines and higher expression of stem cell markers and growth factors compared to adult CDCs. After intramyocardial administration of infant CDCs in the heart of DCM rats, we found that infant CDCs remained in the heart of DCM rats for at least 7 days, improved DCM-induced cardiac function impairment and protected the myocardium by elevating the left ventricular ejection fraction and fraction shortening. However, the effectiveness of transplanted CDCs was reversed later, as increased fibrosis formation instead of angiogenesis was observed. We concluded that infant CDCs, with higher expression of stem cell markers and growth factors, exhibit non-durable heart protection due to limited residence time in the heart of DCM animals, suggesting that multiple administrations of the CDCs or post-regulation after transplantation may be the key for cell therapy in the future.
Collapse
Affiliation(s)
- Siyuan Wang
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Weidan Chen
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Li Ma
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Minghui Zou
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Wenyan Dong
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Haili Yang
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Lei Sun
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China
| | - Xinxin Chen
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.
| | - Jinzhu Duan
- Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China. .,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 JinSui Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
13
|
Zhao ZA, Han X, Lei W, Li J, Yang Z, Wu J, Yao M, Lu XA, He L, Chen Y, Zhou B, Hu S. Lack of Cardiac Improvement After Cardiosphere-Derived Cell Transplantation in Aging Mouse Hearts. Circ Res 2019; 123:e21-e31. [PMID: 30359191 DOI: 10.1161/circresaha.118.313005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Aging is one of the most significant risk factors for cardiovascular diseases, and the incidence of myocardial ischemia increases dramatically with age. Some studies have reported that cardiosphere-derived cells (CDCs) could benefit the injured heart. Nevertheless, the convincing evidence on CDC-induced improvement of aging heart is still limited. OBJECTIVE In this study, we tested whether the CDCs isolated from neonatal mice could benefit cardiac function in aging mice. METHODS AND RESULTS We evaluated cardiac function of PBS- (n=15) and CDC-injected (n=19) aging mice. Echocardiography indicated that left ventricular (LV) ejection fraction (57.46%±3.57% versus 57.86%±2.44%) and LV fraction shortening (30.67%±2.41% versus 30.51%±1.78%) showed similar values in PBS- and CDC-injected mice. The diastolic wall thickness of LV was significantly increased after CDC injection, resulting in reduced diastolic LV volume. The pulse-wave Doppler and tissue Doppler imaging indicated that aging mice receiving PBS or CDC injection presented similar values of the peak early transmitral flow velocity, the peak late transmitral flow velocity, the ratio of the peak early transmitral flow velocity to the peak late transmitral flow velocity, and the ratio of the peak early transmitral flow velocity to the peak early diastolic mitral annular velocity, respectively. Pressure-volume loop experiment indicated that the LV end-diastolic pressure-volume relationship and end-systolic pressure-volume relationship were comparable in both PBS- and CDC-injected mice. Postmortem analysis of aging mouse hearts showed similar fibrotic degree in the 2 groups. In addition, the aging markers showed comparable expression levels in both PBS- and CDC-injected mice. The systemic aging performance measures, including exercise capacity, hair regrowth capacity, and inflammation, showed no significant improvement in CDC-injected mice. Finally, the telomere length was comparable between PBS- and CDC-injected mice. CONCLUSIONS Together, these results indicate that CDCs do not improve heart function and systemic performances in aging mice.
Collapse
Affiliation(s)
- Zhen-Ao Zhao
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Xinglong Han
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Wei Lei
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Jingjing Li
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Zhuangzhuang Yang
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Jie Wu
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Mengchao Yao
- School of Life Science, Shanghai University, China (M.Y.)
| | - Xing-Ai Lu
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Lingjuan He
- the State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (L.H., B.Z.)
| | - Yihuan Chen
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| | - Bin Zhou
- the State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (L.H., B.Z.)
| | - Shijun Hu
- From the Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College (Z.-A.Z., X.H., W.L., J.L., Z.Y., J.W., X.-A.L., Y.C., S.H.), Soochow University, Suzhou, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Medical College (Z.-A.Z., X.H., W.L., J.L., Y.C., S.H.), Soochow University, Suzhou, China
| |
Collapse
|
14
|
Wess G, Wallukat G, Fritscher A, Becker NP, Wenzel K, Müller J, Schimke I. Doberman pinschers present autoimmunity associated with functional autoantibodies: A model to study the autoimmune background of human dilated cardiomyopathy. PLoS One 2019; 14:e0214263. [PMID: 31276517 PMCID: PMC6611557 DOI: 10.1371/journal.pone.0214263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background Autoimmunity associated with autoantibodies against the β1-adrenergic receptor (β1-AAB) is increasingly accepted as the driver of human dilated cardiomyopathy (DCM). Unfortunately, there is a lack of animal models to extend the knowledge about β1-AAB autoimmunity in DCM and to develop appropriate treatment strategies. Objectives To introduce an animal model, we investigated the β1-AAB associated autoimmunity in Doberman Pinscher (DP) with dilated cardiomyopathy, which has similarities to human DCM. Materials and methods Eighty-seven DP with cardiomyopathy in terms of pathological ECG and echocardiography (DoCM) and 31 dogs (at enrollment) without DoCM (controls) were analyzed for serum activity of β1-AAB with a bioassay that records the chronotropic response of spontaneously beating cultured neonatal rat cardiomyocytes to the DP’s IgG. To locate the receptor binding site of β1-AAB and the autoantibody’s sensitivity to inhibition, competing experiments with related blockers were performed with the bioassay. In controls that developed DoCM during follow-up, β1-AAB were analyzed during progress. Results Fifty-nine (67.8%) DoCM dogs and 19 (61.3%) controls were β1-AAB positive. Of the controls that developed DoCM, 8 were β1-AAB positive (p = 0.044 vs. dogs remaining in the control group); their β1-AAB activity increased with the cardiomyopathy progress (p<0.02). To supplement DoCM group with the 9 animals which developed cardiomyopathy in the follow up, a more pronounced β1-AAB positivity became visible in the DoCM group (p = 0.066). Total and cardiac mortality were higher in β1-AAB positive DP (p = 0.002; p = 0037). The dogs’ β1-AAB recognized a specific epitope on the second extracellular receptor and were sensitive to inhibition by drugs already successfully tested to inhibit the corresponding human autoantibody. Conclusions Doberman Pinschers presented β1-AAB associated autoimmunity, similar as in the pathogenesis of human DCM. Consequently, DP could compensate the lack of animal models for the investigation of β1-AAB autoimmunity in human DCM.
Collapse
Affiliation(s)
- Gerhard Wess
- Clinic of Small Animal Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | | | - Anna Fritscher
- Clinic of Small Animal Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | | | | | | | | |
Collapse
|
15
|
Paradies P, Carlucci L, Woitek F, Staffieri F, Lacitignola L, Ceci L, Romano D, Sasanelli M, Zentilin L, Giacca M, Salvadori S, Crovace A, Recchia FA. Intracoronary Gene Delivery of the Cytoprotective Factor Vascular Endothelial Growth Factor-B 167 in Canine Patients with Dilated Cardiomyopathy: A Short-Term Feasibility Study. Vet Sci 2019; 6:vetsci6010023. [PMID: 30845635 PMCID: PMC6466215 DOI: 10.3390/vetsci6010023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disease of dogs and humans characterized by progressive ventricular dilation and depressed contractility and it is a frequent cause of heart failure. Conventional pharmacological therapy cannot reverse the progression of the disease and, in humans, cardiac transplantation remains the only option during the final stages of heart failure. Cytoprotective gene therapy with vascular endothelial growth factor-B167 (VEGF-B167) has proved an effective alternative therapy, halting the progression of the disease in experimental studies on dogs. The aim of this work was to test the tolerability and feasibility of intracoronary administration, under fluoroscopic guidance, of VEGF-B167 carried by adeno-associated viral vectors in canine DCM patients. Ten patients underwent the gene delivery procedure. The intraoperative phase was well tolerated by all dogs. Clinical and echocardiographic assessments at 7- and 30-days post-procedure showed stable conditions compared to the pre-procedure phase. The results of this work indicate that intracoronary VEGF-B167 gene delivery is feasible and tolerated in dogs with DCM. Further monitoring/investigations are ongoing to evaluate the effects of this therapy on disease progression.
Collapse
Affiliation(s)
- Paola Paradies
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Lucia Carlucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, 56100 Pisa, Italy.
| | - Felix Woitek
- Heart Center, Dresden at the Technical University of Dresden, 01067 Dresden, Germany.
| | - Francesco Staffieri
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Luca Lacitignola
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Luigi Ceci
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Daniela Romano
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Mariateresa Sasanelli
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy.
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy.
| | - Stefano Salvadori
- CNR, Institute of Clinical Physiology, Area della Ricerca, 56121 Pisa, Italy.
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, 56100 Pisa, Italy.
| |
Collapse
|
16
|
A missense variant in the titin gene in Doberman pinscher dogs with familial dilated cardiomyopathy and sudden cardiac death. Hum Genet 2019; 138:515-524. [DOI: 10.1007/s00439-019-01973-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/04/2019] [Indexed: 12/30/2022]
|
17
|
Stern JA, Ueda Y. Inherited cardiomyopathies in veterinary medicine. Pflugers Arch 2018; 471:745-753. [PMID: 30284024 DOI: 10.1007/s00424-018-2209-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Abstract
Comparative and translation medicine is of particular value within the field of inherited cardiomyopathies. Despite massive advances in understanding the functional role of mutations in human cardiomyopathies, these advances have frequently failed to translate into medical discoveries that alter patient care. One potential explanation for this failure lies in the lack of suitable translational models that adequately recapitulate human cardiovascular physiology and disease expression. The vast genetic heterogeneity that complicates human cardiomyopathy research is potentially alleviated through the study of naturally occurring large animal models of disease, where incredibly homogenous populations, like those seen in a single breed of dog or cat, may exist (Kol et al., Sci Transl Med 7:308-321, 2015; Ueda and Stern, Yale J Biol Med 90:433-448, 2017). Veterinary medicine is in a unique position to provide research resources and information that may be readily applied to human disease (Kol et al., Sci Transl Med 7:308-321, 2015). Many inherited cardiomyopathies of humans are phenotypically and genotypically similar in veterinary species and ongoing research holds promise for aiding veterinary and human patients alike (Basso et al., Circulation 109:1180-1185, 2004; Fox et al., Cardiovasc Pathol 23:28-34, 2014; Fox et al., Circulation 102:1863-1870, 2000; Kittleson et al., J Vet Cardiol 17 Suppl 1:S53-73, 2015; Ueda and Stern, Yale J Biol Med 90:433-448, 2017). This article presents the current knowledge of inherited cardiomyopathies in dogs, cats, and non-human primates, with a goal of identifying areas of translational research and future directions.
Collapse
Affiliation(s)
- Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA.
| | - Yu Ueda
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA
| |
Collapse
|
18
|
Cardiosphere-derived cells suppress allogeneic lymphocytes by production of PGE2 acting via the EP4 receptor. Sci Rep 2018; 8:13351. [PMID: 30190508 PMCID: PMC6127326 DOI: 10.1038/s41598-018-31569-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022] Open
Abstract
Cardiosphere-derived cells (CDCs) are a cardiac progenitor cell population, which have been shown to possess cardiac regenerative properties and can improve heart function in a variety of cardiac diseases. Studies in large animal models have predominantly focussed on using autologous cells for safety, however allogeneic cell banks would allow for a practical, cost-effective and efficient use in a clinical setting. The aim of this work was to determine the immunomodulatory status of these cells using CDCs and lymphocytes from 5 dogs. CDCs expressed MHC I but not MHC II molecules and in mixed lymphocyte reactions demonstrated a lack of lymphocyte proliferation in response to MHC-mismatched CDCs. Furthermore, MHC-mismatched CDCs suppressed lymphocyte proliferation and activation in response to Concanavalin A. Transwell experiments demonstrated that this was predominantly due to direct cell-cell contact in addition to soluble mediators whereby CDCs produced high levels of PGE2 under inflammatory conditions. This led to down-regulation of CD25 expression on lymphocytes via the EP4 receptor. Blocking prostaglandin synthesis restored both, proliferation and activation (measured via CD25 expression) of stimulated lymphocytes. We demonstrated for the first time in a large animal model that CDCs inhibit proliferation in allo-reactive lymphocytes and have potent immunosuppressive activity mediated via PGE2.
Collapse
|
19
|
Bagno L, Hatzistergos KE, Balkan W, Hare JM. Mesenchymal Stem Cell-Based Therapy for Cardiovascular Disease: Progress and Challenges. Mol Ther 2018; 26:1610-1623. [PMID: 29807782 DOI: 10.1016/j.ymthe.2018.05.009] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/30/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022] Open
Abstract
Administration of mesenchymal stem cells (MSCs) to diseased hearts improves cardiac function and reduces scar size. These effects occur via the stimulation of endogenous repair mechanisms, including regulation of immune responses, tissue perfusion, inhibition of fibrosis, and proliferation of resident cardiac cells, although rare events of transdifferentiation into cardiomyocytes and vascular components are also described in animal models. While these improvements demonstrate the potential of stem cell therapy, the goal of full cardiac recovery has yet to be realized in either preclinical or clinical studies. To reach this goal, novel cell-based therapeutic approaches are needed. Ongoing studies include cell combinations, incorporation of MSCs into biomaterials, or pre-conditioning or genetic manipulation of MSCs to boost their release of paracrine factors, such as exosomes, growth factors, microRNAs, etc. All of these approaches can augment therapeutic efficacy. Further study of the optimal route of administration, the correct dose, the best cell population(s), and timing for treatment are parameters that still need to be addressed in order to achieve the goal of complete cardiac regeneration. Despite significant progress, many challenges remain.
Collapse
Affiliation(s)
- Luiza Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
20
|
Xiang Q, Liao Y, Chao H, Huang W, Liu J, Chen H, Hong D, Zou Z, Xiang AP, Li W. ISL1 overexpression enhances the survival of transplanted human mesenchymal stem cells in a murine myocardial infarction model. Stem Cell Res Ther 2018; 9:51. [PMID: 29482621 PMCID: PMC5828309 DOI: 10.1186/s13287-018-0803-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed as a marker for cardiovascular progenitor cells. This study investigated whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) improves myocardial infarction (MI) treatment outcomes. METHODS The lentiviral vector containing the human elongation factor 1α promoter, which drives the expression of ISL1 (EF1α-ISL1), was constructed using the Multisite Gateway System and used to transduce hMSCs. Flow cytometry, immunofluorescence, Western blotting, TUNEL assay, and RNA sequencing were performed to evaluate the function of ISL1-overexpressing hMSCs (ISL1-hMSCs). RESULTS The in vivo results showed that transplantation of ISL1-hMSCs improved cardiac function in a rat model of MI. Left ventricle ejection fraction and fractional shortening were greater in post-MI hearts after 4 weeks of treatment with ISL1-hMSCs compared with control hMSCs or phosphate-buffered saline. We also found that ISL1 overexpression increased angiogenesis and decreased apoptosis and inflammation. The greater potential of ISL1-hMSCs may be attributable to an increased number of surviving cells after transplantation. Conditioned medium from ISL1-hMSCs decreased the apoptotic effect of H2O2 on the cardiomyocyte cell line H9c2. To clarify the molecular basis of this finding, we employed RNA sequencing to compare the apoptotic-related gene expression profiles of control hMSCs and ISL1-hMSCs. The results showed that insulin-like growth factor binding protein 3 (IGFBP3) was the only gene in ISL1-hMSCs with a RPKM value higher than 100 and that the difference fold-change between ISL1-hMSCs and control hMSCs was greater than 3, suggesting that IGFBP3 might play an important role in the anti-apoptosis effect of ISL1-hMSCs through paracrine effects. Furthermore, the expression of IGFBP3 in the conditioned medium from ISL1-hMSCs was almost fourfold greater than that in conditioned medium from control hMSCs. Moreover, the IGFBP3 neutralization antibody reversed the apoptotic effect of ISL1-hMSCs-CM. CONCLUSIONS These results suggest that overexpression of ISL1 in hMSCs promotes cell survival in a model of MI and enhances their paracrine function to protect cardiomyocytes, which may be mediated through IGFBP3. ISL1 overexpression in hMSCs may represent a novel strategy for enhancing the effectiveness of stem cell therapy after MI.
Collapse
Affiliation(s)
- Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Liao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hua Chao
- Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jia Liu
- Department of Cardiology, the Red Cross hospital of Guangzhou City, the Fourth Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Haixuan Chen
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dongxi Hong
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhengwei Zou
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China. .,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
21
|
Shen D, Shen M, Liang H, Tang J, Wang B, Liu C, Wang P, Dong J, Li L, Zhang J, Caranasos TG. Therapeutic benefits of CD90-negative cardiac stromal cells in rats with a 30-day chronic infarct. J Cell Mol Med 2018; 22:1984-1991. [PMID: 29341439 PMCID: PMC5824400 DOI: 10.1111/jcmm.13517] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/25/2017] [Indexed: 12/18/2022] Open
Abstract
Cardiac stromal cells (CSCs) can be derived from explant cultures, and a subgroup of these cells is viewed as cardiac mesenchymal stem cells due to their expression of CD90. Here, we sought to determine the therapeutic potential of CD90‐positive and CD90‐negative CSCs in a rat model of chronic myocardial infarction. We obtain CD90‐positive and CD90‐negative fractions of CSCs from rat myocardial tissue explant cultures by magnetically activated cell sorting. In vitro, CD90‐negative CSCs outperform CD90‐positive CSCs in tube formation and cardiomyocyte functional assays. In rats with a 30‐day infarct, injection of CD90‐negative CSCs augments cardiac function in the infarct in a way superior to that from CD90‐positive CSCs and unsorted CSCs. Histological analysis revealed that CD90‐negative CSCs increase vascularization in the infarct. Our results suggest that CD90‐negative CSCs could be a development candidate as a new cell therapy product for chronic myocardial infarction.
Collapse
Affiliation(s)
- Deliang Shen
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Miaoda Shen
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Hongxia Liang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Bo Wang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chuang Liu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Peiwen Wang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jianzeng Dong
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.,Department of Cardiology, Beijing Anzhen Hospital of Capital Medical University, Beijing, China
| | - Ling Li
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Dutton LC, Church SAV, Hodgkiss-Geere H, Catchpole B, Huggins A, Dudhia J, Connolly DJ. Cryopreservation of canine cardiosphere-derived cells: Implications for clinical application. Cytometry A 2017; 93:115-124. [PMID: 28834400 DOI: 10.1002/cyto.a.23186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/27/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022]
Abstract
The clinical application of cardiosphere-derived cells (CDCs) to treat cardiac disease has gained increasing interest over the past decade. Recent clinical trials confirm their regenerative capabilities, although much remains to be elucidated about their basic biology. To develop this new treatment modality, in a cost effective and standardized workflow, necessitates the creation of cryopreserved cell lines to facilitate access for cardiac patients requiring urgent therapy. Cryopreservation may however lead to alterations in cell behavior and potency. The aim of this study was to investigate the effect of cryopreservation on canine CDCs. CDCs and mesenchymal stem cells (MSCs) isolated from five dogs were characterized. CDCs demonstrated a population doubling time that was unchanged by cryopreservation (fresh vs. cryopreserved; 57.13 ± 5.27 h vs. 48.94 ± 9.55 h, P = 0.71). This was slower than for MSCs (30.46 h, P < 0.05). The ability to form clones, self-renew, and commit to multiple lineages was unaffected by cryopreservation. Cryopreserved CDCs formed larger cardiospheres compared to fresh cells (P < 0.0001). Fresh CDCs showed a high proportion of CD105+ (89.0% ± 4.98) and CD44+ (99.68% ± 0.13) cells with varying proportions of CD90+ (23.36% ± 9.78), CD34+ (7.18% ± 4.03) and c-Kit+ (13.17% ± 8.67) cells. CD45+ (0.015% ± 0.005) and CD29+ (2.92% ± 2.46) populations were negligible. Increasing passage number of fresh CDCs correlated with an increase in the proportion of CD34+ and a decrease in CD90+ cells (P = 0.003 and 0.03, respectively). Cryopreserved CDCs displayed increased CD34+ (P < 0.001) and decreased CD90+ cells (P = 0.042) when compared to fresh cells. Overall, our study shows that cryopreservation of canine CDCs is feasible without altering their stem characteristics, thereby facilitating their utilization for clinical trials. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Luke C Dutton
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | - Sophie A V Church
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | | | - Brian Catchpole
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK
| | - Anthony Huggins
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK
| | - Jayesh Dudhia
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | - David J Connolly
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| |
Collapse
|
23
|
Dinh PUC, Cores J, Hensley MT, Vandergriff AC, Tang J, Allen TA, Caranasos TG, Adler KB, Lobo LJ, Cheng K. Derivation of therapeutic lung spheroid cells from minimally invasive transbronchial pulmonary biopsies. Respir Res 2017; 18:132. [PMID: 28666430 PMCID: PMC5493087 DOI: 10.1186/s12931-017-0611-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 12/21/2022] Open
Abstract
Background Resident stem and progenitor cells have been identified in the lung over the last decade, but isolation and culture of these cells remains a challenge. Thus, although these lung stem and progenitor cells provide an ideal source for stem-cell based therapy, mesenchymal stem cells (MSCs) remain the most popular cell therapy product for the treatment of lung diseases. Surgical lung biopsies can be the tissue source but such procedures carry a high risk of mortality. Methods In this study we demonstrate that therapeutic lung cells, termed “lung spheroid cells” (LSCs) can be generated from minimally invasive transbronchial lung biopsies using a three-dimensional culture technique. The cells were then characterized by flow cytometry and immunohistochemistry. Angiogenic potential was tested by in-vitro HUVEC tube formation assay. In-vivo bio- distribution of LSCs was examined in athymic nude mice after intravenous delivery. Results From one lung biopsy, we are able to derive >50 million LSC cells at Passage 2. These cells were characterized by flow cytometry and immunohistochemistry and were shown to represent a mixture of lung stem cells and supporting cells. When introduced systemically into nude mice, LSCs were retained primarily in the lungs for up to 21 days. Conclusion Here, for the first time, we demonstrated that direct culture and expansion of human lung progenitor cells from pulmonary tissues, acquired through a minimally invasive biopsy, is possible and straightforward with a three-dimensional culture technique. These cells could be utilized in long-term expansion of lung progenitor cells and as part of the development of cell-based therapies for the treatment of lung diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0611-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Jhon Cores
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Adam C Vandergriff
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA.,Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tyler A Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Thomas G Caranasos
- Divisions of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth B Adler
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Leonard J Lobo
- Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA. .,Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA.
| |
Collapse
|