1
|
Weeratunga P, Harman RM, Jager MC, Van de Walle GR. Footprint-free induced pluripotent stem cells can be successfully differentiated into mesenchymal stromal cells in the feline model. Stem Cell Res Ther 2025; 16:195. [PMID: 40254569 PMCID: PMC12010622 DOI: 10.1186/s13287-025-04325-2] [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: 07/26/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) can propagate indefinitely and give rise to every other cell type, rendering them invaluable for disease modelling, drug development research, and usage in regenerative medicine. While feline iPSCs have been described, there are currently no reports on generating genome integration (footprint)-free iPSCs from domestic cats. Therefore, the objective of this study was to generate feline iPSCs from fetal fibroblasts using non-integrative Sendai virus (SeV) vectors carrying human transcription factors. Moreover, these iPSCs were differentiated into mesenchymal stromal cells (MSCs), which can be used as an alternative to tissue-derived MSCs. METHODS Feline fetal fibroblasts were transduced with CytoTune-iPS 2.0 Sendai Reprogramming vectors at recommended multiplicity of infections (MOI) and cultured for about 6 days. At 7 days post transduction cells were dissociated, replated on inactivated feeder cells and maintained in iPSC medium for 28 days with daily medium change. Emerging iPSC colonies were mechanically passaged and transferred to fresh feeder cells and further passaged every 6-8 days. Four feline iPSC lines were generated, with two selected for further in-depth characterization. Feline iPSCs were then differentiated into MSCs using a serial plating strategy and an inhibitor of the transforming growth factor-β (TGF-β) type I receptor. RESULTS Feline iPSCs exhibited characteristic colony morphology, high nuclear-to-cytoplasmic ratio, positive alkaline phosphatase activity, and expressed feline OCT4, SOX2, and Nanog homeobox (NANOG) stem cell markers. Expression of SeV-derived transgenes decreased during passaging to be eventually lost from the host cells and feline iPSCs could be stably maintained for over 35 passages. Feline iPSCs differentiated into embryoid bodies in vitro and did not form fully differentiated teratomas; instead, they generated in vivo masses containing mesodermal tissue derivatives when injected into immunodeficient mice. Feline iPSC-derived MSCs were plastic adherent, displayed MSC-like morphology, expressed MSC-specific surface markers, and differentiated into cells from the mesodermal lineage in vitro. RNA deep sequencing identified 1,189 differentially expressed genes in feline iPSC-derived MSCs compared to feline iPSCs. CONCLUSION We demonstrated the generation of footprint-free iPSCs from domestic cats and their directed differentiation potential towards MSCs. These SeV-derived feline iPSCs and iPSC-derived MSCs will provide valuable models to study feline diseases and explore novel therapeutic strategies and can serve as translational models for human health, leading to increased knowledge on disease pathogenesis and improved therapeutic interventions.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Mason C Jager
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA.
- Department of Veterinary Pathobiology, The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
2
|
Morawska-Kozłowska M, Pitas M, Zhalniarovich Y. Mesenchymal Stem Cells in Veterinary Medicine-Still Untapped Potential. Animals (Basel) 2025; 15:1175. [PMID: 40282009 PMCID: PMC12024326 DOI: 10.3390/ani15081175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Mesenchymal stem cells (MSCs) hold significant therapeutic potential in veterinary medicine due to their regenerative and immunomodulatory properties. This review examines the clinical applications of MSCs across multiple animal species, including equine, canine, feline, and bovine medicine. MSC therapies have demonstrated promising outcomes in treating musculoskeletal disorders, osteoarthritis, inflammatory diseases, and tissue injuries, particularly in horses and dogs. In cats, MSCs show potential for managing chronic kidney disease, inflammatory bowel disease, and asthma, while in bovine medicine, they offer alternative treatment approaches for mastitis and orthopedic injuries. Despite these advancements, challenges such as treatment standardization, cell sourcing, and potential adverse effects, including tumorigenicity, remain under investigation. The emerging field of MSC-based veterinary medicine highlights its capacity to enhance healing, reduce inflammation, and improve clinical outcomes. However, further research is necessary to optimize treatment protocols and address safety concerns, ensuring the widespread adoption of MSC therapies in veterinary practice.
Collapse
Affiliation(s)
- Magdalena Morawska-Kozłowska
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Mateusz Pitas
- Veterinary Polyclinic, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Yauheni Zhalniarovich
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| |
Collapse
|
3
|
Chen QH, Zheng JY, Wang DC. Asthma and stem cell therapy. World J Stem Cells 2025; 17:103599. [DOI: 10.4252/wjsc.v17.i2.103599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/23/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025] Open
Abstract
The global incidence of asthma, a leading respiratory disorder affecting more than 235 million people, has dramatically increased in recent years. Characterized by chronic airway inflammation and an imbalanced response to airborne irritants, this chronic condition is associated with elevated levels of inflammatory factors and symptoms such as dyspnea, cough, wheezing, and chest tightness. Conventional asthma therapies, such as corticosteroids, long-acting β-agonists, and anti-inflammatory agents, often evoke diverse adverse reactions and fail to reduce symptoms and hospitalization rates over the long term effectively. These limitations have prompted researchers to explore innovative therapeutic strategies, including stem cell-related interventions, offering hope to those afflicted with this incurable disease. In this review, we describe the characteristics of stem cells and critically assess the potential and challenges of stem cell-based therapies to improve disease management and treatment outcomes for asthma and other diseases.
Collapse
Affiliation(s)
- Qiong-Hua Chen
- Department of Respiratory Medicine, Quanzhou Women’s and Children’s Hospital, Clinical Medical College of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Jing-Yang Zheng
- Department of Respiratory Medicine, Quanzhou Women’s and Children’s Hospital, Clinical Medical College of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Da-Chun Wang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Medical School at Houston, Houston, TX 77030, United States
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| |
Collapse
|
4
|
Kwon J, Kim MY, Lee S, Lee J, Yoon HY. Pulmonary passage of canine adipose tissue-derived mesenchymal stem cells through intravenous transplantation in mouse model. J Vet Sci 2024; 25:e36. [PMID: 38834506 PMCID: PMC11156597 DOI: 10.4142/jvs.23300] [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: 12/06/2023] [Revised: 01/16/2024] [Accepted: 03/29/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE The intravenous administration of adipose tissue-derived mesenchymal stem cells (AdMSCs) in veterinary medicine is an attractive treatment option. On the other hand, it can result in severe complications, including pulmonary thromboembolism (PTE). OBJECTIVE The present study assessed the occurrence of PTE after the intravenous infusion of canine AdMSCs (cAdMSCs) into experimental animals. METHODS Five-week-old male BALB/c hairless mice were categorized into groups labeled A to G. In the control group (A), fluorescently stained 2 × 106 cAdMSCs were diluted in 200 μL of suspension and injected into the tail vein as a single bolus. The remaining groups included the following: group B with 5 × 106 cells, group C with 3 × 106 cells, group D with 1 × 106 cells, group E with 1 × 106 cells injected twice with a one-day interval, group F with 2 × 106 cells in 100 μL of suspension, and group G with 2 × 106 cells in 300 μL of suspension. RESULTS Group D achieved a 100% survival rate, while none of the subjects in groups B and C survived (p = 0.002). Blood tests revealed a tendency for the D-dimer levels to increase as the cell dose increased (p = 0.006). The platelet count was higher in the low cell concentration groups and lower in the high cell concentration groups (p = 0.028). A histological examination revealed PTE in most deceased subjects (96.30%). CONCLUSIONS AND RELEVANCE PTE was verified, and various variables were identified as potential contributing factors, including the cell dose, injection frequency, and suspension volume.
Collapse
Affiliation(s)
- Jaeyeon Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Mu-Young Kim
- Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32611, USA
| | - Soojung Lee
- Department of Companion Animal Health, Yeonsung University, Anyang 14011, Korea
| | - Jeongik Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Seoul 05029, Korea
| | - Hun-Young Yoon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
5
|
Barchilon M, Reinero CR. Breathe easy: inhalational therapy for feline inflammatory airway disease. J Feline Med Surg 2023; 25:1098612X231193054. [PMID: 37675792 PMCID: PMC10812033 DOI: 10.1177/1098612x231193054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
PRACTICAL RELEVANCE Feline inflammatory airway diseases, including (but not limited to) asthma, chronic bronchitis and bronchiectasis, are common and incurable disorders. These diseases require lifelong therapy and may result in substantial morbidity and, in some cases, mortality. Goals of therapy include reduction or resolution of clinical signs and the underlying pathologic processes driving those clinical signs. Inhalational therapy has the advantage of topical delivery of drugs to target tissues at higher doses with fewer systemic effects than oral medications. There are multiple options for delivery devices, and proper selection and training on the use of these devices - including acclimation of the cat to the device - can maximize therapeutic efficacy. AIM As inhalational therapy is uncommonly used by many veterinarians and owners, this review article provides a foundation on the selection and use of devices and inhalant medications for specific feline inflammatory airway diseases. Cats present a unique challenge with respect to the use of inhalers, and easy-to-follow steps on acclimating them to the devices are provided. The review also discusses the mechanics of inhalational therapy and helps clarify why certain medications, such as albuterol (salbutamol), fluticasone or budesonide, are chosen for certain diseases. The ultimate aim is that the practitioner should feel more comfortable managing common airway diseases in cats. EVIDENCE BASE In compiling their review, the authors searched the veterinary literature for articles in English that discuss inhalational therapy in cats, and which focus primarily on inhaled glucocorticoids and bronchodilators. While most literature on inhalational therapy in cats is based on experimental feline asthma models, there are some studies demonstrating successful treatment in cats with naturally occurring inflammatory airway disease.
Collapse
Affiliation(s)
- Michael Barchilon
- University of Missouri, College of Veterinary Medicine, Columbia, MO 65211, USA
| | - Carol R Reinero
- University of Missouri, College of Veterinary Medicine, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Armstrong BBS, Pedroso JCM, Conceição Carvalho JD, Ferreira LM. Mesenchymal stem cells in lung diseases and their potential use in COVID-19 ARDS: A systematized review. Clinics (Sao Paulo) 2023; 78:100237. [PMID: 37454534 PMCID: PMC10368758 DOI: 10.1016/j.clinsp.2023.100237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/23/2023] [Accepted: 04/24/2023] [Indexed: 07/18/2023] Open
Abstract
COVID-19 can converge with the pro-inflammatory immunoregulatory mechanisms of chronic lung diseases. Given the disorders inherent to lung transplantation and the inexistence of other definitive therapeutic alternatives, Adipose tissue-derived Stem Cells (ASCs) presented themselves as a therapeutic hope. The purpose of this review is to assess the basis for the potential use of ASCs in lung diseases unresponsive to conventional therapy, relating to their possible use in COVID-19 ARDS. 35 studies comprised this review, 14 being narrative reviews, 19 preclinical trials and two proofs of concept. COVID-19 can converge with the pro-inflammatory immunoregulatory mechanisms of chronic lung diseases. In view of the disorders inherent to lung transplantation and the inexistence of definitive therapeutic alternatives, Adipose tissue-derived Stem Cells (ASCs) presented themselves as a therapeutic hope. Its detailed reading indicated the absence of serious adverse effects and toxicity to the administration of ASCs and suggested possible effectiveness in reducing lung damage, in addition to promoting the recovery of leukocytes and lymphocytes with its immunomodulatory and anti-apoptotic effects. The revised clinical data suggests optimism in the applicability of ASCs in other immunoinflammatory diseases and in severe COVID-19 ARDS. However, further studies are needed to develop a consensus on the methods of collection of ASCs, the ideal dosage schedule, the most effective time and route of administration, as well as on the definition of indications for the administration of ASCs in cases of COVID-19 for conducting clinical trials in near future.
Collapse
Affiliation(s)
| | | | | | - Lydia Masako Ferreira
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Baouche M, Ochota M, Locatelli Y, Mermillod P, Niżański W. Mesenchymal Stem Cells: Generalities and Clinical Significance in Feline and Canine Medicine. Animals (Basel) 2023; 13:1903. [PMID: 37370414 DOI: 10.3390/ani13121903] [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: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells: they can proliferate like undifferentiated cells and have the ability to differentiate into different types of cells. A considerable amount of research focuses on the potential therapeutic benefits of MSCs, such as cell therapy or tissue regeneration, and MSCs are considered powerful tools in veterinary regenerative medicine. They are the leading type of adult stem cells in clinical trials owing to their immunosuppressive, immunomodulatory, and anti-inflammatory properties, as well as their low teratogenic risk compared with pluripotent stem cells. The present review details the current understanding of the fundamental biology of MSCs. We focus on MSCs' properties and their characteristics with the goal of providing an overview of therapeutic innovations based on MSCs in canines and felines.
Collapse
Affiliation(s)
- Meriem Baouche
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Małgorzata Ochota
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Yann Locatelli
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
- Museum National d'Histoire Naturelle, Réserve Zoologique de la Haute Touche, 36290 Obterre, France
| | - Pascal Mermillod
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| |
Collapse
|
8
|
Gholami M, Ghorban K, Sadeghi M, Dadmanesh M, Rouzbahani NH, Dehnavi S. Mesenchymal stem cells and allergic airway inflammation; a therapeutic approach to induce immunoregulatory responses. Int Immunopharmacol 2023; 120:110367. [PMID: 37230032 DOI: 10.1016/j.intimp.2023.110367] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/07/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Allergic airway inflammations are among the essential disorders worldwide that are already considered a significant concern. Mesenchymal stem cells (MSCs) are stromal cells with regenerative potential and immunomodulatory characteristics and are widely administered for tissue repair as an immunoregulatory agent in different inflammatory diseases. The current review summarized primary studies conducted to evaluate the therapeutic potential of MSCs for allergic airway disorders. In this case, modulation of airway pathologic inflammation and infiltration of inflammatory cells were examined, and modulation of the Th1/Th2 cellular balance and humoral responses. Also, the effects of MSCs on the Th17/Treg ratio and inducing Treg immunoregulatory responses along with macrophage and dendritic cell function were evaluated.
Collapse
Affiliation(s)
- Mohammad Gholami
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Medical Microbiology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Khodayar Ghorban
- Department of Immunology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Dadmanesh
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, School Of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Negin Hosseini Rouzbahani
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Immunology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Vientós-Plotts AI, Ericsson AC, McAdams ZL, Rindt H, Reinero CR. Temporal changes of the respiratory microbiota as cats transition from health to experimental acute and chronic allergic asthma. Front Vet Sci 2022; 9:983375. [PMID: 36090168 PMCID: PMC9453837 DOI: 10.3389/fvets.2022.983375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 01/04/2023] Open
Abstract
In humans, deviation from a core airway microbiota may predispose to development, exacerbation, or progression of asthma. We proposed to describe microbiota changes using 16 rRNA sequencing in samples from the upper and lower airways, and rectal swabs of 8 cats after experimental induction of asthma using Bermuda grass allergen, in acute (6 weeks) and chronic (36 weeks) stages. We hypothesized that asthma induction would decrease richness and diversity and alter microbiota composition and structure in the lower airways, without significantly impacting other sites. After asthma induction, richness decreased in rectal (p = 0.014) and lower airway (p = 0.016) samples. B diversity was significantly different between health and chronic asthma in all sites, and between all time points for lower airways. In healthy lower airways Pseudomonadaceae comprised 80.4 ± 1.3% whereas Sphingobacteriaceae and Xanthobacteraceae predominated (52.4 ± 2.2% and 33.5 ± 2.1%, respectively), and Pseudomonadaceae was absent, in 6/8 cats with chronic asthma. This study provides evidence that experimental induction of asthma leads to dysbiosis in the airways and distant sites in both the acute and chronic stages of disease. This article has been published alongside "Respiratory dysbiosis in cats with spontaneous allergic asthma" (1).
Collapse
Affiliation(s)
- Aida I. Vientós-Plotts
- College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Comparative Internal Medicine Laboratory, University of Missouri, Columbia, MO, United States
| | - Aaron C. Ericsson
- College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Zachary L. McAdams
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Hansjorg Rindt
- College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Comparative Internal Medicine Laboratory, University of Missouri, Columbia, MO, United States
| | - Carol R. Reinero
- College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Comparative Internal Medicine Laboratory, University of Missouri, Columbia, MO, United States
| |
Collapse
|
10
|
Mukherjee P, Roy S, Ghosh D, Nandi SK. Role of animal models in biomedical research: a review. Lab Anim Res 2022; 38:18. [PMID: 35778730 PMCID: PMC9247923 DOI: 10.1186/s42826-022-00128-1] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
The animal model deals with the species other than the human, as it can imitate the disease progression, its’ diagnosis as well as a treatment similar to human. Discovery of a drug and/or component, equipment, their toxicological studies, dose, side effects are in vivo studied for future use in humans considering its’ ethical issues. Here lies the importance of the animal model for its enormous use in biomedical research. Animal models have many facets that mimic various disease conditions in humans like systemic autoimmune diseases, rheumatoid arthritis, epilepsy, Alzheimer’s disease, cardiovascular diseases, Atherosclerosis, diabetes, etc., and many more. Besides, the model has tremendous importance in drug development, development of medical devices, tissue engineering, wound healing, and bone and cartilage regeneration studies, as a model in vascular surgeries as well as the model for vertebral disc regeneration surgery. Though, all the models have some advantages as well as challenges, but, present review has emphasized the importance of various small and large animal models in pharmaceutical drug development, transgenic animal models, models for medical device developments, studies for various human diseases, bone and cartilage regeneration model, diabetic and burn wound model as well as surgical models like vascular surgeries and surgeries for intervertebral disc degeneration considering all the ethical issues of that specific animal model. Despite, the process of using the animal model has facilitated researchers to carry out the researches that would have been impossible to accomplish in human considering the ethical prohibitions.
Collapse
Affiliation(s)
- P Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - S Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - D Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - S K Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India.
| |
Collapse
|
11
|
Creamer DG, Schmiedt CW, Bullington AC, Caster CM, Schmiedt JM, Hurley DJ, Berghaus RD. Influence of exposure to microbial ligands, immunosuppressive drugs and chronic kidney disease on endogenous immunomodulatory gene expression in feline adipose-derived mesenchymal stem cells. J Feline Med Surg 2022; 24:e43-e56. [PMID: 35302413 PMCID: PMC11104253 DOI: 10.1177/1098612x221083074] [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] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Feline autologous mesenchymal stem cells (MSCs) show promise for immunomodulatory activity, but the functional impact of chronic kidney disease (CKD), concurrent immunosuppressive drug administration or infection is unknown. The study objectives compare endogenous cytokine gene expression (interleukin [IL]-6, IL-10, IL-12p40, IL-18 and transforming growth factor beta [TGF-β]) in adipose-derived MSCs (aMSCs) from cats with and without CKD, following in vitro exposure to microbial ligands and treatment with common immunosuppressive drugs. METHODS Previously obtained aMSCs, phenotype CD44+, CD90+, CD105+ and MHCII-, from cats with (n = 6) and without (n = 6) CKD were compared via real-time PCR (RT-PCR) for immunomodulatory gene expression. aMSCs were exposed in vitro to lipopolysaccharide (LPS), peptidoglycan or polyinosinic:polycytidylic acid (Poly I:C), simulating bacterial or viral exposure, respectively. aMSCs were also exposed to ciclosporin, dexamethasone or methotrexate. Gene expression was measured using RT-PCR, and Cq was utilized after each run to calculate the delta cycle threshold. RESULTS aMSCs isolated from healthy and CKD cats showed no significant differences in gene expression in the five measured cytokines. No significant changes in measured gene expression after drug treatment or microbial ligand stimulation were observed between normal or CKD affected cats. Proinflammatory genes (IL-6, IL-12p40 and IL-18) showed altered expression in aMSCs from both groups when compared with the same cells in standard culture after exposure to methotrexate. Poly I:C altered IL-6 and TGF-β gene expression in aMSCs from both healthy and CKD cats when compared with the same cells in standard culture. CONCLUSIONS AND RELEVANCE The five genes tested showed no statistical differences between aMSCs from healthy or CKD cats. There was altered cytokine gene expression between the control and treatment groups of both healthy and CKD cats suggesting feline aMSCs have altered function with immunosuppressive treatment or microbial ligand exposure. Although the current clinical relevance of this pilot study comparing brief exposure to select agents in vitro in aMSCs from a small number of cats is unknown, the study highlights a need for continued investigation into the effects of disease and concurrent therapies on use of cell-based therapies in feline patients.
Collapse
Affiliation(s)
- Danielle G Creamer
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Chad W Schmiedt
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Anna Claire Bullington
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Courtney M Caster
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jennifer M Schmiedt
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - David J Hurley
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Roy D Berghaus
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
12
|
Villatoro AJ, Martín-Astorga MDC, Alcoholado C, Kazantseva L, Cárdenas C, Fariñas F, Becerra J, Visser R. Secretory Profile of Adipose-Tissue-Derived Mesenchymal Stem Cells from Cats with Calicivirus-Positive Severe Chronic Gingivostomatitis. Viruses 2022; 14:v14061146. [PMID: 35746618 PMCID: PMC9228153 DOI: 10.3390/v14061146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/14/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023] Open
Abstract
The feline calicivirus (FCV) causes infections in cats all over the world and seems to be related to a broad variety of clinical presentations, such as feline chronic gingivostomatitis (FCGS), a severe oral pathology in cats. Although its etiopathogeny is largely unknown, FCV infection is likely to be a main predisposing factor for developing this pathology. During recent years, new strategies for treating FCGS have been proposed, based on the use of mesenchymal stem cells (MSC) and their regenerative and immunomodulatory properties. The main mechanism of action of MSC seems to be paracrine, due to the secretion of many biomolecules with different biological functions (secretome). Currently, several pathologies in humans have been shown to be related to functional alterations of the patient’s MSCs. However, the possible roles that altered MSCs might have in different diseases, including virus-mediated diseases, remain unknown. We have recently demonstrated that the exosomes produced by the adipose-tissue-derived MSCs (fAd-MSCs) from cats suffering from FCV-positive severe and refractory FCGS showed altered protein contents. Based on these findings, the goal of this work was to analyze the proteomic profile of the secretome produced by feline adipose-tissue-derived MSCs (fAd-MSCs) from FCV-positive patients with FCGS, in order to identify differences between them and to increase our knowledge of the etiopathogenesis of this disease. We used high-resolution mass spectrometry and functional enrichment analysis with Gene Ontology to compare the secretomes produced by the fAd-MSCs of healthy and calicivirus-positive FCGS cats. We found that the fAd-MSCs from cats with FCGS had an increased expression of pro-inflammatory cytokines and an altered proteomic profile compared to the secretome produced by cells from healthy cats. These findings help us gain insight on the roles of MSCs and their possible relation to FCGS, and may be useful for selecting specific biomarkers and for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Antonio J. Villatoro
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
- Grupo Ynmun, Inmunología Clínica y Terapia Celular (IMMUNESTEM), 29071 Málaga, Spain
| | - María del Carmen Martín-Astorga
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
| | - Liliya Kazantseva
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
| | - Casimiro Cárdenas
- Research Support Central Services (SCAI) of the University of Málaga, 29071 Málaga, Spain;
| | - Fernando Fariñas
- Grupo Ynmun, Spanish Association for the Research in Immunological and Infectious Diseases, 29071 Málaga, Spain;
| | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rick Visser
- Laboratory of Bioengineering and Tissue Regeneration, Department of Cell Biology, Genetics and Physiology, Biomedical Research Institute of Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain; (A.J.V.); (M.d.C.M.-A.); (C.A.); (L.K.); (J.B.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-952-131-858
| |
Collapse
|
13
|
Isolation and Characterization of Cat Olfactory Ecto-Mesenchymal Stem Cells. Animals (Basel) 2022; 12:ani12101284. [PMID: 35625130 PMCID: PMC9137790 DOI: 10.3390/ani12101284] [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: 03/23/2022] [Revised: 04/28/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Cat’s health is impacted by several diseases and lesions for which cell therapy could be an interesting treatment. Mesenchymal stem cells or adult stem cells are found in developed tissue. Olfactory mucosa contains stem cells called olfactory ecto-mesenchymal stem cells which have already been isolated from various animals as dogs and horses. The aim of this study was to evaluate the feasibility of collecting olfactory ecto-mesenchymal stem cells in cats. For that purpose, four cats were biopsied; the cells were collected and characterized. They show stemness features and differentiation capabilities as all the other mammals previously studied. Therefore, olfactory ecto-mesenchymal stem cells could be a promising tool for feline regenerative medicine. Abstract The olfactory mucosa contains olfactory ecto-mesenchymal stem cells (OE-MSCs) which show stemness features, multipotency capabilities, and have a therapeutic potential. The OE-MSCs have already been collected and isolated from various mammals. The aim of this study was to evaluate the feasibility of collecting, purifying and amplifying OE-MSCs from the cat nasal cavity. Four cats were included in the study. Biopsies of olfactory mucosa were performed on anesthetized animals. Then, the olfactory OE-MSCs were isolated, and their stemness features as well as their mesodermal differentiation capabilities were characterized. Olfactory mucosa biopsies were successfully performed in all subjects. From these biopsies, cellular populations were rapidly generated, presenting various stemness features, such as a fibroblast-like morphology, nestin and MAP2 expression, and sphere and colony formation. These cells could differentiate into neural and mesodermal lineages. This report shows for the first time that the isolation of OE-MSCs from cat olfactory mucosa is possible. These cells showed stemness features and multilineage differentiation capabilities, indicating they may be a promising tool for autologous grafts and feline regenerative medicine.
Collapse
|
14
|
Taghizadeh S, Keyhanmanesh R, Rahbarghazi R, Rezaie J, Delkhosh A, Hassanpour M, Heiran H, Ghaffari-Nasab A, Ahmadi M. Systemic administration of c-Kit + cells diminished pulmonary and vascular inflammation in rat model of chronic asthma. BMC Mol Cell Biol 2022; 23:11. [PMID: 35209844 PMCID: PMC8876378 DOI: 10.1186/s12860-022-00410-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To circumvent some pitfalls related to acute status, chronic model of asthma is conceived to be more suitable approach to guarantee the conditions which are similar to human pulmonary disease. Here, possible therapeutic mechanisms were monitored by which c-kit+ bone marrow cells can attenuate vascular inflammation in rat model of chronic asthma. RESULTS Data revealed c-Kit+ cells could significantly reduce pathological injures in asthmatic rats via modulating the expression of IL-4, INF-γ, ICAM-1 and VCAM-1 in lung tissues and TNF-α, IL-1β and NO levels in BALF (p < 0.001 to p < 0.05). Besides, c-Kit+ cells reduced increased levels of VCAM-1 evaluated by immunohistochemistry staining. In contrast to c-Kit+ cells, c-Kit- cells could not exert beneficial effects in the asthmatic conditions. CONCLUSION Overall, we found that systemic administration of C-kit positive cells can diminish pulmonary and vascular inflammation of chronic asthmatic changes in a rat model. These cells are eligible to suppress inflammation and nitrosative stress in lung tissue coincides with the reduction of pathological changes. These data indicate that C-kit positive cells be used as an alternative cell source for the amelioration of asthmatic changes.
Collapse
Affiliation(s)
- Sajjad Taghizadeh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Aref Delkhosh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Heiran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Kim EY, Kim HS, Hong KS, Chung HM, Park SP, Noh G. Mesenchymal stem/stromal cell therapy in atopic dermatitis and chronic urticaria: immunological and clinical viewpoints. Stem Cell Res Ther 2021; 12:539. [PMID: 34635172 PMCID: PMC8503727 DOI: 10.1186/s13287-021-02583-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/30/2021] [Indexed: 12/29/2022] Open
Abstract
Allergic diseases are immune-mediated diseases. Allergies share a common immunopathogenesis, with specific differences according to the specific disease. Mesenchymal stem/stromal cells (MSCs) have been applied to people suffering from allergic and many other diseases. In this review, the immunologic roles of MSCs are systemically reviewed according to disease immunopathogenesis from a clinical viewpoint. MSCs seem to be a promising therapeutic modality not only as symptomatic treatments but also as causative and even preventive treatments for allergic diseases, including atopic dermatitis and chronic urticaria.
Collapse
Affiliation(s)
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science, The Graduate School of Dong-A University, Busan, Korea.,Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | | | - Hyung-Min Chung
- Miraecellbio Co., Ltd., Seoul, Korea.,Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Se-Pill Park
- Miraecellbio Co., Ltd., Seoul, Korea. .,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, 63243, Korea.
| | - Geunwoong Noh
- Department of Allergy, Allergy and Clinical Immunology Center, Cheju Halla General Hospital, Doreongno 65, Jeju-si, 63127, Jeju Special Self-Governing Province, Korea.
| |
Collapse
|
16
|
Taechangam N, Walker NJ, Borjesson DL. Feline adipose-derived mesenchymal stem cells induce effector phenotype and enhance cytolytic function of CD8+ T cells. Stem Cell Res Ther 2021; 12:495. [PMID: 34488876 PMCID: PMC8420035 DOI: 10.1186/s13287-021-02558-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Feline adipose-derived mesenchymal stem cells (ASCs) engage with a variety of immune cells and have been used in several clinical trials for the treatment of inflammatory and immune-dysregulated diseases in cats, but the impact they exert on the functional characteristics on T cells, particularly CD8+ T cells, remains to be elucidated. METHODS Modified mixed leukocyte reaction was performed between feline ASCs and PBMCs. Changes of cell cycle stages, phenotype and cellular senescence were determined through flow cytometry and gene expression analysis. Cytotoxicity assay was performed to evaluate CD8+ T cell effector function. RESULTS Feline ASCs induce cell cycle arrest on CD8+ T cells in a contact-dependent manner, downregulate CD8 surface expression, and shift their phenotype toward terminally differentiated effector cells (CD57+, CD45R+, CD62L-). CD8 T cells interacted with feline ASCs also upregulated granzyme B, IL-2 and KLRG-1 expression and have enhanced cytotoxic potential, evident by the increased percentage of lysis on target cells. CONCLUSIONS Our findings suggest that feline ASCs (1) alter CD8+ T cells toward terminally differentiated, proinflammatory effector phenotype with limited proliferative capacity, and (2) enhance their cytotoxic potential through granzyme B upregulation. These cytotoxic CD8+ T cells could aid in disease cure in cases caused by an underlying, unresolved viral infection.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, University of California, Vet Med 3A, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology and Immunology, University of California, Vet Med 3A, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology and Immunology, University of California, Vet Med 3A, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
17
|
Park SG, An JH, Li Q, Chae HK, Park SM, Lee JH, Ahn JO, Song WJ, Youn HY. Feline adipose tissue-derived mesenchymal stem cells pretreated with IFN-γ enhance immunomodulatory effects through the PGE₂ pathway. J Vet Sci 2021; 22:e16. [PMID: 33774932 PMCID: PMC8007449 DOI: 10.4142/jvs.2021.22.e16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/12/2020] [Accepted: 01/07/2021] [Indexed: 01/03/2023] Open
Abstract
Background Preconditioning with inflammatory stimuli is used to improve the secretion of anti-inflammatory agents in stem cells from variant species such as mouse, human, and dog. However, there are only few studies on feline stem cells. Objectives This study aimed to evaluate the immune regulatory capacity of feline adipose tissue-derived (fAT) mesenchymal stem cells (MSCs) pretreated with interferon-gamma (IFN-γ). Methods To assess the interaction of lymphocytes and macrophages with IFN-γ-pretreated fAT-MSCs, mouse splenocytes and RAW 264.7 cells were cultured with the conditioned media from IFN-γ-pretreated MSCs. Results Pretreatment with IFN-γ increased the gene expression levels of cyclooxygenase-2, indoleamine 2,3-dioxygenase, hepatocyte growth factor, and transforming growth factor-beta 1 in the MSCs. The conditioned media from IFN-γ-pretreated MSCs increased the expression levels of M2 macrophage markers and regulatory T-cell markers compared to those in the conditioned media from naive MSCs. Further, prostaglandin E2 (PGE2) inhibitor NS-398 attenuated the immunoregulatory potential of MSCs, suggesting that the increased PGE2 levels induced by IFN-γ stimulation is a crucial factor in the immune regulatory capacity of MSCs pretreated with IFN-γ. Conclusions IFN-γ pretreatment improves the immune regulatory profile of fAT-MSCs mainly via the secretion of PGE2, which induces macrophage polarization and increases regulatory T-cell numbers.
Collapse
Affiliation(s)
- Seol Gi Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Ju Hyun An
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Qiang Li
- Department of Veterinary Medicine, College of Agriculture, Yanbian University, Yanji, Jilin 133000, China
| | - Hyung Kyu Chae
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Su Min Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jeong Hwa Lee
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jin Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Woo Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute of Veterinary Science, Jeju National University, Jeju 63243, Korea.
| | - Hwa Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
18
|
Brennan LC, O’Sullivan A, MacLoughlin R. Cellular Therapy for the Treatment of Paediatric Respiratory Disease. Int J Mol Sci 2021; 22:ijms22168906. [PMID: 34445609 PMCID: PMC8396271 DOI: 10.3390/ijms22168906] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Respiratory disease is the leading cause of death in children under the age of 5 years old. Currently available treatments for paediatric respiratory diseases including bronchopulmonary dysplasia, asthma, cystic fibrosis and interstitial lung disease may ameliorate symptoms but do not offer a cure. Cellular therapy may offer a potential cure for these diseases, preventing disease progression into adulthood. Induced pluripotent stem cells, mesenchymal stromal cells and their secretome have shown great potential in preclinical models of lung disease, targeting the major pathological features of the disease. Current research and clinical trials are focused on the adult population. For cellular therapies to progress from preclinical studies to use in the clinic, optimal cell type dosage and delivery methods need to be established and confirmed. Direct delivery of these therapies to the lung as aerosols would allow for lower doses with a higher target efficiency whilst avoiding potential effect of systemic delivery. There is a clear need for research to progress into the clinic for the treatment of paediatric respiratory disease. Whilst research in the adult population forms a basis for the paediatric population, varying disease pathology and anatomical differences in paediatric patients means a paediatric-centric approach must be taken.
Collapse
Affiliation(s)
- Laura C. Brennan
- College of Medicine, Nursing & Health Sciences, National University of Ireland, H91 TK33 Galway, Ireland;
| | - Andrew O’Sullivan
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, H91 HE94 Galway, Ireland;
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, H91 HE94 Galway, Ireland;
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Correspondence:
| |
Collapse
|
19
|
Proteomic Analysis of the Secretome and Exosomes of Feline Adipose-Derived Mesenchymal Stem Cells. Animals (Basel) 2021; 11:ani11020295. [PMID: 33498940 PMCID: PMC7912403 DOI: 10.3390/ani11020295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The enormous advances in stem cell research have generated high expectations in the development of new therapies to repair or regenerate damaged tissues. For this reason, laboratory studies of stem cells enable scientists to learn about cells’ essential properties. Specifically, in recent years, therapies based on mesenchymal stem cells have become an interesting alternative for the treatment of different complex pathologies in veterinary medicine. Mesenchymal stem cells secrete a wide variety of therapeutic elements such as bioactive molecules and extracellular vesicles (e.g., exosomes). Thus, it is essential to characterize them before future use as biotechnological products. Therefore, the objective of this study was to determine and compare their protein profile to understand better the mechanisms of action of these components and facilitate their possible use in future therapies. The data demonstrate the existence of different proteins responsible for the biological effects of cells. In addition, these approaches and techniques can contribute to the better prediction of clinical outcomes of mesenchymal stem cell treatment. Abstract Mesenchymal stem cells (MSCs) have been shown to have therapeutic efficacy in different complex pathologies in feline species. This effect is attributed to the secretion of a wide variety of bioactive molecules and extracellular vesicles, such as exosomes, with significant paracrine activity, encompassed under the concept of the secretome. However, at present, the exosomes from feline MSCs have not yet been studied in detail. The objective of this study is to analyze and compare the protein profiles of the secretome as a whole and its exosomal fraction from feline adipose-derived MSCs (fAd-MSCs). For this, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Protein–Protein Interaction Networks Functional Enrichment Analysis (STRING) were utilized. A total of 239 proteins were identified in the secretome, and 228 proteins specific to exosomes were identified, with a total of 133 common proteins. The proteins identified in the secretome were located in the extracellular regions and in the cytoplasm, while the exosomal proteins were located mainly in the membrane, cytoplasm and cytosol. Regarding function, in the secretome, proteins involved in different metabolic pathways, in pathways related to the immune system and the endocrine system and in the processing of proteins in the endoplasmic reticulum predominated. In contrast, proteins specific to exosomes were predominantly associated with endocytosis, cell junctions, platelet activation and other cell signaling pathways. The possible future use of the secretome, or some of its components, such as exosomes, would provide a non-cell-based therapeutic strategy for the treatment of different diseases that would avoid the drawbacks of cell therapy.
Collapse
|
20
|
Mirershadi F, Ahmadi M, Rezabakhsh A, Rajabi H, Rahbarghazi R, Keyhanmanesh R. Unraveling the therapeutic effects of mesenchymal stem cells in asthma. Stem Cell Res Ther 2020; 11:400. [PMID: 32933587 PMCID: PMC7493154 DOI: 10.1186/s13287-020-01921-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
Asthma is a chronic inflammatory disease associated with airway hyper-responsiveness, chronic inflammatory response, and excessive structural remodeling. The current therapeutic strategies in asthmatic patients are based on controlling the activity of type 2 T helper lymphocytes in the pulmonary tissue. However, most of the available therapies are symptomatic and expensive and with diverse side outcomes in which the interruption of these modalities contributes to the relapse of asthmatic symptoms. Up to date, different reports highlighted the advantages and beneficial outcomes regarding the transplantation of different stem cell sources, and relevant products from for the diseases' alleviation and restoration of injured sites. However, efforts to better understand by which these cells elicit therapeutic effects are already underway. The precise understanding of these mechanisms will help us to translate stem cells into the clinical setting. In this review article, we described current knowledge and future perspectives related to the therapeutic application of stem cell-based therapy in animal models of asthma, with emphasis on the underlying therapeutic mechanisms.
Collapse
Affiliation(s)
- Fatemeh Mirershadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran.,Department of Physiology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Rajabi
- Koc University Research Center for Translational Medicine (KUTTAM), Koc University School of Medicine, Istanbul, Turkey.,Department of Pulmonary Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51548-53431, Iran.
| | - Rana Keyhanmanesh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran. .,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Abstract
Regenerative medicine is a complex field of research, with much hope placed on the ability to use regenerative therapies such as stem cells to provide new treatment options for many frustrating disease processes in human and veterinary medicine. Significant research is still needed and ongoing from basic mechanistic studies to advanced bioengineering applications to practical cell delivery methods. Small studies of mesenchymal stem cell therapy shows significant promise in inflammatory feline diseases. Continued research will hopefully help determine the potential of mesenchymal stem cell therapy in feline medicine and lead to development of safe and effective products for clinical use.
Collapse
Affiliation(s)
- Tracy L Webb
- Clinical Sciences, Colorado State University, 1678 Campus Delivery, Fort Collins, CO 80523, USA.
| |
Collapse
|
22
|
Safety and immunomodulatory properties of equine peripheral blood-derived mesenchymal stem cells in healthy cats. Vet Immunol Immunopathol 2020; 227:110083. [PMID: 32563854 DOI: 10.1016/j.vetimm.2020.110083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Due to the immunomodulatory properties of mesenchymal stem cells (MSCs) through stimulation of endogenous immune cells by paracrine signals and cell contact, they have been proposed as alternative treatment option for many inflammatory and immune-mediated diseases in veterinary medicine. However, the long-term cultivation possibilities of feline MSCs are currently compromised due to a restricted proliferation capacity. Therefore, the xenogeneic use of equine peripheral blood-derived MSCs (ePB-MSCs) would present an interesting alternative thanks to their superior cultivation properties. To the authors' knowledge, there are currently no safety reports concerning the xenogeneic use of ePB-MSCs in cats. Therefore, the overall goal of this preliminary study was to investigate if ePB-MSCs can safely be administered in healthy cats and by extension evaluating their immunogenic and immunomodulatory properties. METHODS Ten healthy cats were intravenously (i.v.) injected with 3 × 105 ePB-MSCs at three time points (T0, T1, T2). All cats were daily inspected by the caretaker and underwent a physical examination with hematological and biochemical analysis at day 0 (T0), week 2 (T1), week 4 (T2) and week 6 (T3) by a veterinarian. Furthermore, a modified mixed lymphocyte reaction (MLR) was performed at T0 and T3 for each cat in order to evaluate immunogenic and immunomodulatory properties of the ePB-MSCs RESULTS: No adverse clinical effects could be detected following repeated i.v. administration of ePB-MSCs in all cats. Significant lower protein (T1: P-value = 0.002; T2: P-value > 0.001; T3: P-value = 0.004) and albumin levels (T1: P-value = 0.003; T2: P-value = 0.001) were seen after repeated administration of ePB-MSCs, compared to T0. However, all biochemical and hematological parameters stayed within clinical acceptance level. In addition, the repeated injections did not induce a cellular immune response before and after repeated ePB-MSCs administration. Furthermore, convincing immunomodulatory properties of ePB-MSCs on feline peripheral blood mononuclear cells were confirmed in the MLR-assay CONCLUSION: This preliminary study demonstrates that ePB-MSCs can safely be administered in healthy cats and provide a promising alternative for the treatment of various inflammatory diseases in cats.
Collapse
|
23
|
Akkoç T, Genç D. Asthma immunotherapy and treatment approaches with mesenchymal stem cells. Immunotherapy 2020; 12:665-674. [PMID: 32489107 DOI: 10.2217/imt-2019-0194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Asthma is a chronic inflammatory disease of the airways where exaggerated T helper 2 immune responses and inflammatory mediators play a role. Current asthma treatment options can effectively suppress symptoms and control the inflammatory process; however, cannot modulate the dysregulated immune response. Allergen-specific immunotherapy is one of the effective treatments capable of disease modification. Injecting allergens under the skin in allergen-specific immunotherapy can reduce asthma and improve the sensitivity of the lungs, however, has a risk of severe reactions. Mesenchymal stem cells have immunoregulatory activity with their soluble mediators and contact dependent manner. In this review, we focus on the current treatment strategies with mesenchymal stem cells in asthma as a new therapeutic tool and compare those with immunotherapy.
Collapse
Affiliation(s)
- Tunç Akkoç
- Department of Pediatric Allergy & Immunology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Deniz Genç
- Department of Pediatric Health & Diseases, Faculty of Health Sciences, Muğla Sıtkı Koçman University, Muğla, Turkey
| |
Collapse
|
24
|
Fujimoto Y, Yokozeki T, Yokoyama A, Tabata Y. Basic fibroblast growth factor enhances proliferation and hepatocyte growth factor expression of feline mesenchymal stem cells. Regen Ther 2020; 15:10-17. [PMID: 32490062 PMCID: PMC7256438 DOI: 10.1016/j.reth.2020.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/26/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction The objective of this study is to evaluate the effect of basic fibroblast growth factor (bFGF) on the proliferation and secretion activity of feline adipose-derived mesenchymal stem cells (MSC). Methods Feline MSC isolated from the subcutaneous adipose tissue of cats were cultured with or without bFGF. Results The bFGF addition enhanced the proliferation of feline MSC to a significant great extent compared with that without bFGF, although the cell proliferation tended to increase with the bFGF concentration. In addition, adipogenic and osteogenic staining assay demonstrated that the bFGF addition allowed MSC to maintain the differentiation ability even after the proliferation. Moreover, no change in the surface markers of MSC was observed between the cultures with or without bFGF. A quantitative RT-PCR assay revealed that the HGF and TSG-6 expression significantly increased by the bFGF addition. The highest mRNA expression of MMP-2 was observed for cells cultured in 1000 ng/ml bFGF concentration. Conclusions The culture with bFGF is a promising way to enhance the proliferation, and HGF secretion ability of MSC as well as maintain their differentiation ability and immunophenotype nature. Feline adipose-derived mesenchymal stem cells (MSC) was cultured with or without the basic fibroblast growth factor (bFGF). The bFGF enhanced the proliferation and increased the mRNA expression of HGF, TSG-6, and MMP-2. The bFGF addition was not influenced to the differentiation ability and cell surface marker of MSC.
Collapse
Key Words
- Basic fibroblast growth factor
- CKD, chronic kidney disease
- ECM, extracellular matrix
- FBS, fetal bovine serum
- FGF, basic fibroblast growth factor
- Feline
- GAPDH, gliyceraldehyde-3-phosphate dehydrogenase
- HGF, hepatocyte growth factor
- Hepatocyte growth factor
- MMP-2, matrix metalloproteinase-2
- MSC, mesenchymal stem cells
- Mesenchymal stem cell
- P1, passage 1
- Proliferation
- SVF, stromal vascular fraction
- TSG-6, tumor necrosis factor-stimulated gene 6
- Tumor necrosis factor-stimulated gene 6
Collapse
Affiliation(s)
- Youhei Fujimoto
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan.,Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
25
|
MacDonald ES, Barrett JG. The Potential of Mesenchymal Stem Cells to Treat Systemic Inflammation in Horses. Front Vet Sci 2020; 6:507. [PMID: 32039250 PMCID: PMC6985200 DOI: 10.3389/fvets.2019.00507] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
One hallmark of mesenchymal stem cells (MSCs) is the ability to differentiate into multiple tissue types which assists in tissue regeneration. Another hallmark of MSCs is their potent anti-inflammatory and immunomodulatory properties and the potential to treat inflammatory, immune-mediated, and ischemic conditions. In equine practice, MSCs have shown efficacy in the treatment of musculoskeletal disorders such as tendinopathy, meniscal tears and cartilage injury. However, there are many equine disease processes and conditions that may benefit from the immunomodulatory properties of MSCs. Examples include conditions associated with overwhelming acute inflammatory response such as systemic inflammatory response syndrome to chronic diseases characterized by a prolonged low level of inflammation such as equine asthma and recurrent uveitis. For the acute inflammatory response processes, there is often high morbidity and mortality with no effective immunomodulatory treatment to prevent the overwhelming synthesis of proinflammatory mediators. For chronic inflammatory disease processes, frequently long-term corticosteroid treatment is the therapeutic mainstay, with serious potential complications. Thus, there is an unmet need for alternative anti-inflammatory treatments for both acute and chronic illnesses in horses. While MSCs show promise for such conditions, much research is needed before a clinically safe and effective treatment will be available. Optimal MSC tissue source, patient vs. donor source (autologous vs. allogeneic) and cell growth conditions need to be determined for each problem. For immediate use, allogeneic MSC treatments is preferable, but immune tolerance and adequate safety require further study. MSC collection and cryopreservation from horses before they are injured or ill, whether from umbilical cord tissue, bone marrow or adipose might become more widespread. Once these fundamental approaches to treating specific diseases with MSCs are determined, the route of administration, dose and timing of administration also need to be studied. To provide a framework for development of MSC immunomodulatory treatments, this article reviews the current understanding of equine MSC anti-inflammatory and immunomodulatory properties and proposes how MSC therapy may be further developed to treat acute onset systemic inflammatory processes and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth S MacDonald
- Marion duPont Scott Equine Medical Center, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, VA, United States
| | - Jennifer G Barrett
- Marion duPont Scott Equine Medical Center, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, VA, United States
| |
Collapse
|
26
|
Li H, Tian Y, Xie L, Liu X, Huang Z, Su W. Mesenchymal stem cells in allergic diseases: Current status. Allergol Int 2020; 69:35-45. [PMID: 31445840 DOI: 10.1016/j.alit.2019.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Allergic diseases, which include asthma, allergic skin diseases, allergic rhinitis and allergic conjunctivitis, have already garnered worldwide public health attention over recent decades. Mesenchymal stem cells (MSCs) have gradually emerged as a potential method for treating allergic diseases due to their immunosuppressive characteristics, tissue repair ability and secretion of various biological factors. This potential of MSC-based therapy has been confirmed in clinical and preclinical studies, which report the therapeutic benefits of MSCs for various allergic diseases and explore the antiallergic mechanisms. In this review, we focus on the discoveries and biological mechanisms of MSCs as a therapeutic tool in allergic diseases. We discuss the challenges of conducting MSC studies as well as future directions.
Collapse
|
27
|
Abstract
Asthma is an important allergic lower-airway disease in cats affecting approximately 1% to 5% of the pet cat population. New diagnostics are being developed to help better differentiate asthma from other lower-airway diseases and improve monitoring. In addition, new treatments are being developed to help in refractory cases or in those cases in which traditional therapeutics are contraindicated. This article discusses potential pitfalls in the diagnosis of asthma. In addition, current literature investigating new diagnostic tests and therapies for feline asthma is reviewed.
Collapse
Affiliation(s)
- Julie E Trzil
- IndyVet Emergency and Specialty Hospital, 5425 Victory Drive, Indianapolis, IN 46203, USA.
| |
Collapse
|
28
|
Dias IE, Pinto PO, Barros LC, Viegas CA, Dias IR, Carvalho PP. Mesenchymal stem cells therapy in companion animals: useful for immune-mediated diseases? BMC Vet Res 2019; 15:358. [PMID: 31640767 PMCID: PMC6805418 DOI: 10.1186/s12917-019-2087-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells are multipotent cells, with capacity for self-renewal and differentiation into tissues of mesodermal origin. These cells are possible therapeutic agents for autoimmune disorders, since they present remarkable immunomodulatory ability.The increase of immune-mediated diseases in veterinary medicine has led to a growing interest in the research of these disorders and their medical treatment. Conventional immunomodulatory drug therapy such as glucocorticoids or other novel therapies such as cyclosporine or monoclonal antibodies are associated with numerous side effects that limit its long-term use, leading to the need for developing new therapeutic strategies that can be more effective and safe.The aim of this review is to provide a critical overview about the therapeutic potential of these cells in the treatment of some autoimmune disorders (canine atopic dermatitis, feline chronic gingivostomatitis, inflammatory bowel disease and feline asthma) compared with their conventional treatment.Mesenchymal stem cell-based therapy in autoimmune diseases has been showing that this approach can ameliorate clinical signs or even cause remission in most animals, with the exception of canine atopic dermatitis in which little to no improvement was observed.Although mesenchymal stem cells present a promising future in the treatment of most of these disorders, the variability in the outcomes of some clinical trials has led to the current controversy among authors regarding their efficacy. Mesenchymal stem cell-based therapy is currently requiring a deeper and detailed analysis that allows its standardization and better adaptation to the intended therapeutic results, in order to overcome current limitations in future trials.
Collapse
Affiliation(s)
- Inês Esteves Dias
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
| | - Pedro Olivério Pinto
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
- Coimbra University Veterinary Hospital, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
| | - Luís Carlos Barros
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
| | - Carlos Antunes Viegas
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães Portugal
- ICVS/3B’s – PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Isabel Ribeiro Dias
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães Portugal
- ICVS/3B’s – PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Pedro Pires Carvalho
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
- Vetherapy, 479 St, San Francisco, CA 94103 USA
| |
Collapse
|
29
|
Panopoulos I, Auriemma E, Specchi S, Diana A, Pietra M, Papastefanou A, Zini E, Cipone M. 64-multidetector CT anatomical assessment of the feline bronchial and pulmonary vascular structures. J Feline Med Surg 2019; 21:893-901. [PMID: 30407139 PMCID: PMC11132249 DOI: 10.1177/1098612x18807778] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of the study was to provide a detailed anatomical study of the feline bronchial and vascular structures by using CT angiography (CTA). METHODS Adult cats with no respiratory clinical signs were enrolled in a CTA protocol to provide an anatomical study of the thorax. The dimensions, number of branches and branching pattern (monopodial vs dichotomic) of both bronchial and pulmonary vascular structures were evaluated under positive inspiration apnoea. A linear generalised estimating equations analysis (Spearman's rho) was used to identify statistical correlation between tracheal diameter, age and body weight of the cats. RESULTS Fourteen cats met the inclusion criteria. The pulmonary arteries had larger diameters than the pulmonary veins, and the pulmonary veins had larger diameters than the bronchial structures. A higher number of segmental bronchial and pulmonary vascular branches was observed in the left caudal lung lobe than in the other lobes. The monopodial branching pattern of both bronchial and pulmonary vascular structures was predominant in all cats of our study (100%) in cranial, caudal and right middle lung lobes, while a dichotomic branching pattern of the bronchial and pulmonary vascular structures of the accessory lung lobe was seen in 13 cats (93%). Thirteen cats (93%) had three pulmonary vein ostia, and one cat (7%) also presented with an additional left intermediate pulmonary vein ostium. Variation in the number of segmental pulmonary vein branches was noted in the right caudal lung lobe. There was no statistical correlation between tracheal diameter, age and weight. CONCLUSIONS AND RELEVANCE Architecture of the feline bronchovascularr structures belongs to a mixed type of monopodial and dichotomic branching pattern. In cats, the pulmonary venous drainage system predominately presents three pulmonary vein ostia. Variations in the type of formation and the number of branches of the pulmonary venous drainage system were noted.
Collapse
Affiliation(s)
| | - Edoardo Auriemma
- Department of Diagnostic Imaging, Veterinary Institute of Novara, Novara, Italy
| | - Swan Specchi
- Department of Diagnostic Imaging, Veterinary Institute of Novara, Novara, Italy
| | - Alessia Diana
- Department of Veterinary Medical Sciences, University of Bologna, Italy
| | - Marco Pietra
- Department of Veterinary Medical Sciences, University of Bologna, Italy
| | | | - Eric Zini
- Clinic for Small Animal Internal Medicine, Zurich University, Vetsuisse-Faculty, Zurich, Switzerland
- Department of Animal Medicine, Production and Health, University of Padua, Legnaro, Italy
- Department of Internal Medicine, Istituto Veterinario di Novara, Novara, Italy
| | - Mario Cipone
- Department of Veterinary Medical Sciences, University of Bologna, Italy
| |
Collapse
|
30
|
Thoracic computed tomographic interpretation for clinicians to aid in the diagnosis of dogs and cats with respiratory disease. Vet J 2019; 253:105388. [PMID: 31685132 DOI: 10.1016/j.tvjl.2019.105388] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 02/08/2023]
Abstract
In humans, high-resolution computed tomography (CT) is a key diagnostic modality for pulmonary disorders. Its success likely lies in excellent correlation of lung diseases with associated subgross anatomic changes, as assessed by histopathology, and because of a multidisciplinary approach between clinicians, radiologists and pathologists. Although thoracic CT studies have been performed in dogs and cats for nearly three decades, there is a lack of uniformity in both protocols for acquisition and in terminology used to describe lesions. Importantly, terms such as a bronchial, interstitial, and alveolar patterns are inappropriate descriptors for canine and feline thoracic CT imaging changes; instead, lung patterns should be classified as increased or decreased attenuation, nodular patterns, and linear patterns, with specific vocabulary to describe subtypes of lesions. In this manuscript, the authors provide an overview of basic CT principles, strategies to optimize and acquire high-quality diagnostic studies (inclusive of paired inspiratory and expiratory series, contrast and triphasic angiography) and provide a roadmap for systematic interpretation of thoracic CT images.
Collapse
|
31
|
Lin SC, Liou YM, Ling TY, Chuang YH, Chiang BL. Placenta-Derived Mesenchymal Stem Cells Reduce the Interleukin-5 Level Experimentally in Children with Asthma. Int J Med Sci 2019; 16:1430-1438. [PMID: 31673233 PMCID: PMC6818200 DOI: 10.7150/ijms.33590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/21/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) have been investigated as a new treatment option for various diseases in recent years. However, the role of placenta-derived MSCs in children with asthma remains unclear. We assessed the effect of placenta-derived MSCs on T cell immune responses and cytokine IL-5 levels according to cultures in children with and without asthma. Study design: We enrolled children with and without asthma and recorded asthma symptom scores in the asthma group. Blood samples from children were collected to isolate peripheral blood mononuclear cells (PBMCs) and determine the total IgE level. The PBMCs were cultured in vitro with or without MSCs after stimulation with human anti-CD3 and anti-CD28 antibodies (0.5 μg/mL) to evaluate the effect of placenta-derived MSCs. Flow cytometry was performed to detect the activation and proliferation of CD4+ and CD8+ T cells. Pre- and post-culture IL-5 levels were measured in all samples. Results: The percentages of activation and proliferation among CD4+ and CD8+ T cells after coculture with MSCs were significantly lower in the asthma group (P < 0.05). IL-5 levels differed significantly between the PBMC culture and PBMC + MSC (P+S) coculture in the asthma group (P < 0.05). IL-5 levels differed significantly between the PBMC culture and P+S coculture in both the lower (P < 0.05) and higher (P < 0.0005) IgE asthma subgroups. IL-5 levels were also decreased in children with all severities of asthma (P < 0.05). Conclusions: Placenta-derived MSCs exerted an anti-IL-5 effect and reduced the IL-5 level in culture in different subgroups of children with asthma.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yih-Mei Liou
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Hui Chuang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
32
|
Garrity S, Lee-Fowler T, Reinero C. Feline asthma and heartworm disease: Clinical features, diagnostics and therapeutics. J Feline Med Surg 2019; 21:825-834. [PMID: 31446863 PMCID: PMC10814146 DOI: 10.1177/1098612x18823348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PRACTICAL RELEVANCE For feline practitioners, the cat with a cough or respiratory distress and thoracic radiographs with a bronchial or bronchointerstitial pattern suggests lower airway disease. Two important differentials, allergic asthma and heartworm disease (HWD), have many overlapping clinicopathologic features, but also clear and important differences in terms of cause and disease progression, treatment and prognosis. Notably, asthma is readily treatable and HWD is preventable. CLINICAL SIGNIFICANCE Feline HWD comprises two clinical syndromes: the comparatively recently described heartworm-associated respiratory disease (HARD) and adult HWD. The former is much more common; very few cats with HARD develop adult HWD. In HARD, following death of immature worms, pulmonary lesions may improve over time ('self-cure'). Lesions of adult HWD also improve over time as long as reinfection does not occur; however, with death of adult heartworms, mortality is high, and the prognosis is guarded. In asthma, morbidity is relatively high, but mortality is low, with an overall good to excellent prognosis. DISEASE RECOGNITION Feline asthma is encountered worldwide. In the authors' impression, feline HWD is often under-recognized. AIMS The aim of this review is to assist clinicians in differentiating feline asthma from feline HWD; as such, the emphasis is on distinguishing clinical features, as well as on diagnostics, therapy and prognosis. In differentiating these conditions, clinicians can attempt the goal of properly managing these diseases and can best educate owners on prognosis. EVIDENCE BASE For both feline asthma and feline HWD, the authors have drawn on the available peer-reviewed literature studies involving experimental models as well as spontaneous disease.
Collapse
Affiliation(s)
- Sarah Garrity
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| | - Tekla Lee-Fowler
- Department of Clinical Sciences, Auburn University, Auburn, AL, USA
| | - Carol Reinero
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| |
Collapse
|
33
|
Taechangam N, Iyer SS, Walker NJ, Arzi B, Borjesson DL. Mechanisms utilized by feline adipose-derived mesenchymal stem cells to inhibit T lymphocyte proliferation. Stem Cell Res Ther 2019; 10:188. [PMID: 31238978 PMCID: PMC6593543 DOI: 10.1186/s13287-019-1300-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/20/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Feline adipose-derived mesenchymal stem cells (ASCs) have been successfully used in clinical trials for the treatment of immune-mediated diseases with T cell dysregulation. However, the immunomodulatory pathways utilized by feline ASCs to suppress T cell activation have not been fully determined. We investigated the mechanisms used by feline ASCs to inhibit T cell proliferation, including the soluble factors and the cell-cell contact ligands responsible for ASC-T cell interaction. METHODS The immunomodulatory activity of feline ASCs was evaluated via cell cycle analysis and in vitro mixed leukocyte reaction using specific immunomodulatory inhibitors. Cell-cell interactions were assessed with static adhesion assays, also with inhibitors. RESULTS Feline ASCs decrease T cell proliferation by causing cell cycle arrest in G0-G1. Blocking prostaglandin (PGE2), but not IDO, partially restored lymphocyte proliferation. Although PDL-1 and CD137L are both expressed on activated feline ASCs, only the interaction of intercellular adhesion molecule 1 (ICAM-1, CD54) with its ligand, lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), was responsible for ASC-T cell adhesion. Blocking this interaction reduced cell-cell adhesion and mediator (IFN-γ) secretion and signaling. CONCLUSIONS Feline ASCs utilize PGE2 and ICAM-1/LFA-1 ligand interaction to inhibit T cell proliferation with a resultant cell cycle arrest in G0-G1. These data further elucidate the mechanisms by which feline ASCs interact with T cells, help define appropriate T cell-mediated disease targets in cats that may be amenable to ASC therapy, and may also inform potential translational models for human diseases.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Smita S Iyer
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, University of California, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA. .,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
34
|
Feline Adult Adipose Tissue-Derived Multipotent Stromal Cell Isolation and Differentiation. Methods Mol Biol 2019; 1773:173-188. [PMID: 29687390 DOI: 10.1007/978-1-4939-7799-4_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The cat, as a species, is somewhat new to the field of adult multipotent stromal cells. Despite the relative phylogenetic distance between the domestic cat, Felis silvestris catus, and humans, they share some similar health challenges like diabetes, kidney disease and asthma. There is a plethora of current investigative efforts focused on adult adipose tissue-derived multipotent stromal cell (ASC) therapies to address these and other conditions. Given the small size of domestic cats, particular attention to optimize cell isolation from relatively little tissue is a necessary condition of feline ASC studies and therapies. Additionally, there are some unique features of culture conditions to test and confirm feline ASC plasticity. This chapter contains a few of the novel aspects of feline ASC isolation and culture.
Collapse
|
35
|
Zhang LB, He M. Effect of mesenchymal stromal (stem) cell (MSC) transplantation in asthmatic animal models: A systematic review and meta-analysis. Pulm Pharmacol Ther 2018; 54:39-52. [PMID: 30496803 DOI: 10.1016/j.pupt.2018.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/17/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Over the years, mesenchymal stromal (stem) cells (MSCs) have been pre-clinically applied in the treatment of variety kinds of diseases including asthma and chronic lung diseases. Aim of the current study was to systematically review and to conduct meta-analysis on the published studies of MSC treatment in asthma animal models. METHODS Publications on the MSC and asthma treatment was thoroughly searched in the electronic databases. Statistical analysis was then performed using the Comprehensive Meta-Analysis software (Version 3). Effect of MSC therapy on asthma model was assessed by Hedges's g with 95% confidence intervals (95% CIs). Random effect model was used due to the heterogeneity between the studies. RESULTS Meta-analysis of the 32 included studies showed that MSC transplantation was significantly in favor of attenuating lung injury and remodeling (Hedges's g = -9.104 ± 0.951 with 95% CI: -10.969 ∼ -7.240, P < 0.001) and airway inflammation (Hedges's g = -4.146 ± 0.688 with 95% CI: -5.495 ∼ -2.797, P < 0.001). The mechanism of MSC therapy in asthma seems to be regulating the balance of Th1 cytokine and Th2 cytokines (IFN-γ: Hedges's g = 4.779 ± 1.408 with 95% CI: 1.099-2.725, P < 0.001; IL-4: Hedges's g = -10.781 ± 1.062 with 95% CI: -12.863 ∼ -8.699, P < 0.001; IL-5: Hedges's g = -10.537 ± 1.269 with 95% CI: -13.025 ∼ -8.050, P < 0.001; IL-13: Hedges's g = -6.773 ± 0.788 with 95% CI: -8.318 ∼ -5.229, P < 0.001). CONCLUSION Findings of the current systemic review suggested a potential role for MSCs in asthma treatment although it is still challenging in clinical practice. The mechanisms of MSCs in pre-clinical asthma treatment may be associated with attenuating airway inflammation through regulating Th1 and Th2 cytokines.
Collapse
Affiliation(s)
- Li-Bo Zhang
- Department of Respiratory Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min He
- Department of Respiratory Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
36
|
Hooi KS, Defarges AM, Sanchez AL, Nykamp SG, Weese JS, Abrams-Ogg ACG, Bienzle D. Comparison of bronchoscopic and nonbronchoscopic bronchoalveolar lavage in healthy cats. Am J Vet Res 2018; 79:1209-1216. [DOI: 10.2460/ajvr.79.11.1209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Rahbarghazi R, Keyhanmanesh R, Aslani MR, Hassanpour M, Ahmadi M. Bone marrow mesenchymal stem cells and condition media diminish inflammatory adhesion molecules of pulmonary endothelial cells in an ovalbumin-induced asthmatic rat model. Microvasc Res 2018; 121:63-70. [PMID: 30343002 DOI: 10.1016/j.mvr.2018.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Although excitements related to stem cell therapeutic outcomes have been highlighted enormously in asthma, the vast majority of works were conducted by researchers in animal models. Elucidating the mechanisms underlying the therapeutic effects of MSCs in asthmatic rats will provide a rational basis for assuring maximal safety of future clinical application of stem cells. In the current study, we sought to investigate the possible paracrine mechanism by which direct injection of MSCs and/or CM attenuate efficiently Th2-mediated inflammation in asthmatic lung tissues with the focus on ICAM-1 and VCAM-1 expression. METHODS Male rats were divided into four experimental groups (n = 6); healthy rats received PBS intratracheally (group C), sensitized rats received PBS intratracheally (group S), sensitized rats received CM intratracheally (group S + CM), and sensitized rats received PBS intratracheally containing 2 × 106 rBMMSCs (group S + MSCs). Two weeks post-transplantation, the expression of interleukin (IL)-5, -12 and INF-γ, ICAM-1 and VCAM-1 were assessed along with pathological injuries and the homing of MSCs into the lung tissues. RESULTS Our results showed CM, and notably rBMMSCs, returned the expression of IL-5, IL-12, INF-γ, ICAM-1, and VCAM-1 (p < 0.001 to p < 0.05) to the normal levels. Based on data, pathological injuries in pulmonary specimens of asthmatic rats were significantly attenuated (p < 0.001 to p < 0.05). Moreover, rBMMSCs had potential to successfully home to an asthmatic niche in cell-administrated rats. CONCLUSIONS Our data noted the potency of CM and especially MSCs in ameliorating pathological changes via intra-tracheal route presumably by targeting ICAM-1 and VCAM-1 in lung tissues in rat asthma model.
Collapse
Affiliation(s)
- Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Aslani
- Ardabil Imam Khomeini Educational and Clinical Hospital, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mehdi Hassanpour
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Lee BY, Li Q, Song WJ, Chae HK, Kweon K, Ahn JO, Youn HY. Altered properties of feline adipose-derived mesenchymal stem cells during continuous in vitro cultivation. J Vet Med Sci 2018; 80:930-938. [PMID: 29669964 PMCID: PMC6021870 DOI: 10.1292/jvms.17-0563] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cytotherapy with mesenchymal stem cells (MSCs) has been studied in many species, and
often requires in vitro cell expansion to obtain therapeutic doses of
stem cells. Because the characteristics of MSCs, such as self-renewal and multi-lineage
differentiation, can be altered by long-term culture, it is important to maintain stemness
during cultivation. This study assessed the changes in the characteristics of feline
adipose tissue-derived (fAT)-MSCs during in vitro passaging. Stem cells
isolated from the adipose tissue of donor cats were cultured for seven sub-passages.
Proliferation capacity was analyzed by calculating the cell doubling time and by
colorimetric assay. Expression of stem cell-specific markers was evaluated by quantitative
reverse transcription (qRT)-PCR and immunophenotyping. Expression of adipogenic and
osteogenic differentiation markers was also measured by qRT-PCR. Histochemical staining
and measurement of β-galactosidase activity were conducted to detect cellular senescence.
The cell proliferation rate decreased significantly at passage 5 (P5). Gene expression
levels of pluripotency markers (Sox2, Nanog and
Klf4) and stem cell surface markers (CD9,
CD44, CD90 and CD105) decreased
during continuous culture; in most assays, statistically significant changes were observed
at P5. The ability of cells to undergo adipogenic or osteogenic differentiation was
inversely proportional to the number of passages. The proportion of senescent cells
increased with the number of passages. These results suggest that repeated passages alter
the proliferation and multipotency of fAT-MSCs. In clinical trials, early-passage cells
should be used to achieve the maximum therapeutic effect.
Collapse
Affiliation(s)
- Bo-Yeon Lee
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Woo-Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung-Kyu Chae
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyeong Kweon
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin-Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
39
|
Kim HR, Lee J, Byeon JS, Gu NY, Lee J, Cho IS, Cha SH. Extensive characterization of feline intra-abdominal adipose-derived mesenchymal stem cells. J Vet Sci 2018; 18:299-306. [PMID: 27456770 PMCID: PMC5639082 DOI: 10.4142/jvs.2017.18.3.299] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/15/2016] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) isolated from various tissues have been well characterized for therapeutic application to clinical diseases. However, in contrast to MSCs from other animal species, the characteristics of feline MSCs have not been fully documented. In this study, we conducted extensive characterization of feline adipose tissue-derived MSCs (fAD-MSCs). Study fAD-MSCs were individually isolated from the intra-abdominal adipose tissues of six felines. The expression levels of cell surface markers and pluripotent markers were evaluated. Next, proliferation capacity was analyzed by performing cumulative population doubling level (CPDL) and doubling time (DT) calculation assays. Differentiation potentials of fAD-MSCs into mesodermal cell lineages were analyzed by examining specific staining and molecular markers. All fAD-MSCs positively expressed cell surface markers such as CD29, CD44, CD90, CD105, CD166, and MHC-I, while CD14, CD34, CD45, and CD73 were negatively expressed. The CPDL of the fAD-MSCs was maintained until passage 5 to 6 (P5 to P6), whereas DT increased after P3 to P4. Also, stem cell-specific pluripotent markers (Oct3/4, Nanog, and SSEA-4) were detected. Importantly, all fAD-MSCs demonstrated mesodermal differentiation capacity. These results suggest that fully characterized fAD-MSCs could be beneficial when considering the use of these cells in feline disease research.
Collapse
Affiliation(s)
- Hee-Ryang Kim
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Jeong Su Byeon
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Jiyun Lee
- Kangnam Animal Hospital, Pyeongtaek 17982, Korea
| | - In-Soo Cho
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Sang-Ho Cha
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| |
Collapse
|
40
|
Villatoro AJ, Claros S, Fernández V, Alcoholado C, Fariñas F, Moreno A, Becerra J, Andrades JA. Safety and efficacy of the mesenchymal stem cell in feline eosinophilic keratitis treatment. BMC Vet Res 2018; 14:116. [PMID: 29587744 PMCID: PMC5870249 DOI: 10.1186/s12917-018-1413-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/06/2018] [Indexed: 01/07/2023] Open
Abstract
Background Feline eosinophilic keratitis (FEK) is a chronic keratopathy caused by a suspected immune mediated response to an unknown antigenic stimulus. The purpose of this study was to investigate the safety and therapeutic effects of allogeneic feline adipose-derived mesenchymal stromal cells (fAd-MSCs) implanted subconjunctival around the ocular surface lesion in five cats with FEK refractory to current available treatments. Results FEK was diagnosed by clinical appearance and evidence of eosinophil and/or mast cells in corneal cytology. Each animal was treated with two applications of 2 × 106 million of fAd-MSCs 2 months apart. Ocular surface integrity was assessed before treatment and at 1, 3, 6 and 11 months after treatment. Clinical signs showed a significant change during the follow-up with resolution of the corneal and conjunctiva lesions and there were no signs of regression or worsening. Conclusions Implanted cells were well-tolerated and effective reducing clinical signs of FEK with a sustained effect during the study period. None of the animals showed systemic or local complications during the study. To our knowledge, this is the first time in literature that local implantation of allogeneic fAd-MSCs has been found as an effective therapeutic alternative to treat cats with FEK.
Collapse
Affiliation(s)
- Antonio J Villatoro
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Silvia Claros
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Viviana Fernández
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Fernando Fariñas
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Antonio Moreno
- Hospital veterinario Alhaurín el Grande. Alhaurín el Grande, 29120, Málaga, Spain
| | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.,Laboratory of Bioengineering and Tissue Regeneration, Andalusian Center for Nanomedicine and Biotechnology-BIONAND, 29590, Málaga, Spain
| | - José A Andrades
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain. .,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.
| |
Collapse
|
41
|
Quimby JM, Borjesson DL. Mesenchymal stem cell therapy in cats: Current knowledge and future potential. J Feline Med Surg 2018; 20:208-216. [PMID: 29478398 PMCID: PMC10816289 DOI: 10.1177/1098612x18758590] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Practical relevance: Stem cell therapy is an innovative field of scientific investigation with tremendous potential for clinical application in veterinary medicine. Based on the known desirable immunomodulatory properties of mesenchymal stem cells, this therapy holds promise for the treatment of a variety of inflammatory diseases in cats. AIMS This review details our current understanding of feline stem cell biology and proposed mechanism of action. Studies performed in feline clinical trials for diseases including gingivostomatitis, chronic enteropathy, asthma and kidney disease are summarized, with the goal of providing an overview of the current status of this treatment modality and its potential for the future.
Collapse
Affiliation(s)
- Jessica M Quimby
- The Ohio State University, Department of Veterinary Clinical Sciences, Columbus, OH 43210, USA
| | - Dori L Borjesson
- University of California–Davis, Veterinary Institute for Regenerative Cures, and Department of Pathology, Microbiology and Immunology, Davis, CA 95616, USA
| |
Collapse
|
42
|
Malaquias MAS, Oyama LA, Jericó PC, Costa I, Padilha G, Nagashima S, Lopes-Pacheco M, Rebelatto CLK, Michelotto PV, Xisto DG, Brofman PRS, Rocco PRM, de Noronha L. Effects of mesenchymal stromal cells play a role the oxidant/antioxidant balance in a murine model of asthma. Allergol Immunopathol (Madr) 2018; 46:136-143. [PMID: 29174587 DOI: 10.1016/j.aller.2017.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/30/2017] [Accepted: 06/21/2017] [Indexed: 01/10/2023]
Abstract
Asthma is a heterogeneous disease characterised by chronic airway inflammation. One of the most devastating consequences of this inflammatory process is the generation of reactive oxygen and nitrogen species responsible for oxidative stress. The aim of this study is to analyse the efficiency of treatment with human bone marrow-derived mesenchymal stromal cells (hMSC) in maintaining the oxidative balance in a murine model of allergic asthma by quantifying nitrotyrosine in lung tissues. After confirmation of asthma in the experimental model, samples of lung parenchyma were submitted to immunohistochemical assessment. Intravenous administration of hMSC reduced the levels of nitrotyrosine in the ASTHMA-hMSC group compared to those in the ASTHMA-SAL group. In conclusion, therapeutic administration of hMSC had a beneficial effect on oxidative stress, reducing the levels of nitrotyrosine in lung tissues in a model of allergic asthma.
Collapse
Affiliation(s)
- M A S Malaquias
- Pathology Experimental Laboratory, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil.
| | - L A Oyama
- Pathology Experimental Laboratory, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - P C Jericó
- Pathology Experimental Laboratory, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - I Costa
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - G Padilha
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - S Nagashima
- Pathology Experimental Laboratory, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - M Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - C L K Rebelatto
- Experimental Laboratory for Cell Culture, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - P V Michelotto
- Experimental Laboratory for Cell Culture, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - D G Xisto
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P R S Brofman
- Experimental Laboratory for Cell Culture, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - P R M Rocco
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - L de Noronha
- Pathology Experimental Laboratory, School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| |
Collapse
|
43
|
Yu HC, Chiang BL. Toll-like receptor 2 ligation of mesenchymal stem cells alleviates asthmatic airway inflammation. J Allergy Clin Immunol 2018; 142:284-287.e5. [PMID: 29382594 DOI: 10.1016/j.jaci.2017.12.996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 12/06/2017] [Accepted: 12/20/2017] [Indexed: 12/01/2022]
Affiliation(s)
- Hui-Chieh Yu
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei City, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei City, Taiwan.
| |
Collapse
|
44
|
Takeda K, Webb TL, Ning F, Shiraishi Y, Regan DP, Chow L, Smith MJ, Ashino S, Guth AM, Hopkins S, Gelfand EW, Dow S. Mesenchymal Stem Cells Recruit CCR2 + Monocytes To Suppress Allergic Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 200:1261-1269. [PMID: 29352000 DOI: 10.4049/jimmunol.1700562] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 12/03/2017] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSC) exert immune modulatory properties and previous studies demonstrated suppressive effects of MSC treatment in animal models of allergic airway inflammation. However, the underlying mechanisms have not been fully elucidated. We studied the role of MSC in immune activation and subsequent recruitment of monocytes in suppressing airway hyperresponsiveness and airway inflammation using a mouse model of allergic airway inflammation. MSC administration prior to or after allergen challenge inhibited the development of airway inflammation in allergen-sensitized mice. This was accompanied by an influx of CCR2-positive monocytes, which were localized around injected MSC in the lungs. Notably, IL-10-producing monocytes and/or macrophages were also increased in the lungs. Systemic administration of liposomal clodronate or a CCR2 antagonist significantly prevented the suppressive effects of MSC. Activation of MSC by IFN-γ leading to the upregulation of CCL2 expression was essential for the suppressive effects, as administration of wild-type MSC into IFN-γ-deficient recipients, or IFN-γ receptor-deficient or CCL2-deficient MSC into wild-type mice failed to suppress airway inflammation. These results suggest that MSC activation by IFN-γ, followed by increased expression of CCL2 and recruitment of monocytes to the lungs, is essential for suppression by MSC in allergen-induced airway hyperresponsiveness and airway inflammation.
Collapse
Affiliation(s)
- Katsuyuki Takeda
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Tracy L Webb
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Fangkun Ning
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Yoshiki Shiraishi
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Daniel P Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Lyndah Chow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Mia J Smith
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Shigeru Ashino
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Amanda M Guth
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Sophie Hopkins
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Erwin W Gelfand
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|
45
|
Kitoko JZ, de Castro LL, Nascimento AP, Abreu SC, Cruz FF, Arantes AC, Xisto DG, Martins MA, Morales MM, Rocco PRM, Olsen PC. Therapeutic administration of bone marrow-derived mesenchymal stromal cells reduces airway inflammation without up-regulating Tregs in experimental asthma. Clin Exp Allergy 2017; 48:205-216. [PMID: 29068567 DOI: 10.1111/cea.13048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 09/08/2017] [Accepted: 10/18/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Prophylactic administration of mesenchymal stromal cells (MSCs) derived from adipose (AD-MSC) and bone marrow tissue (BM-MSC) in ovalbumin-induced asthma hinders inflammation in a Treg-dependent manner. It is uncertain whether MSCs act through Tregs when inflammation is already established in asthma induced by a clinically relevant allergen. OBJECTIVE Evaluate the effect of therapeutic administration of MSCs on inflammation and Treg cells in house dust mite (HDM)-induced asthma. METHODS BM-MSCs and AD-MSCs were administered intratracheally to C57BL/6 mice 1 day after the last HDM challenge. Lung function, remodelling and parenchymal inflammation were assayed 3 or 7 days after MSCs treatment, through invasive plethysmography and histology, respectively. Bronchoalveolar lavage fluid (BALF) and mediastinal lymph nodes (mLNs) were assessed regarding the inflammatory profile by flow cytometry, ELISA and qRT-PCR. MSCs were studied regarding their potential to induce Treg cells from primed and unprimed lymphocytes in vitro. RESULTS BM-MSCs, but not AD-MSCs, reduced lung influx of eosinophils and B cells and increased IL-10 levels in HDM-challenged mice. Neither BM-MSCs nor AD-MSCs reduced lung parenchymal inflammation, airway hyperresponsiveness or mucus hypersecretion. BM-MSCs and AD-MSCs did not up-regulate Treg cell counts within the airways and mLNs, but BM-MSCs decreased the pro-inflammatory profile of alveolar macrophages. Co-culture of BM-MSCs and AD-MSCs with allergen-stimulated lymphocytes reduced Treg cell counts in a cell-to-cell contact-independent manner, although co-culture of both MSCs with unprimed lymphocytes up-regulated Treg cell counts. CONCLUSIONS MSCs therapeutically administered exert anti-inflammatory effects in the airway of HDM-challenged mice, but do not ameliorate lung function or remodelling. Although MSC pre-treatment can increase Treg cell numbers, it is highly unlikely that the MSCs will induce Treg cell expansion when lymphocytes are allergenically primed in an established lung inflammation.
Collapse
Affiliation(s)
- J Z Kitoko
- Laboratory of Clinical Bacteriology and Immunology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - L L de Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A P Nascimento
- Laboratory of Clinical Bacteriology and Immunology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - S C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - F F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A C Arantes
- Laboratory of Inflammation, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - D G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - M A Martins
- Laboratory of Inflammation, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - M M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Olsen
- Laboratory of Clinical Bacteriology and Immunology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
46
|
Zajic LB, Webb TL, Webb P, Coy JW, Dow SW, Quimby JM. Comparison of proliferative and immunomodulatory potential of adipose-derived mesenchymal stem cells from young and geriatric cats. J Feline Med Surg 2017; 19:1096-1102. [PMID: 27913779 PMCID: PMC11110994 DOI: 10.1177/1098612x16680703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Objectives The objective of this study was to compare the ability of adipose-derived mesenchymal stem cells (aMSCs) generated from young vs geriatric cats to proliferate in culture, suppress lymphocyte proliferation and undergo senescence. Methods Adipose tissues from five young (<5 years) and six geriatric (>10 years) cats were harvested and cryopreserved for subsequent aMSC isolation and culture. aMSC proliferation in culture was compared via determination of time until passage two and by 3-(4,5-demethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. The immunomodulatory capacity of aMSCs was assessed using lymphocyte proliferation assays, and senescence was evaluated using senescence-associated B-galactosidase (SABG) expression. All assays were performed on aMSCs between passage two and passage three. Results aMSCs from geriatric cats took significantly longer ( P = 0.008) to reach passage two (median 11 days, range 9-22 days) compared with aMSCs from young healthy cats (median 7 days, range 6-8 days). No significant difference was detected between young and geriatric cats in terms of their ability to suppress lymphocyte proliferation. SABG expression was not significantly different between young and geriatric aMSCs. Conclusions and relevance Compared with young feline aMSCs, geriatric aMSCs are significantly impaired in their ability to rapidly proliferate to passage two following initial culture, presenting a concern for autologous therapy. Nonetheless, once the cells are expanded, young and geriatric cat aMSCs appear to be equivalent in terms of their ability to functionally suppress T-cell activation and proliferation.
Collapse
Affiliation(s)
- Lara B Zajic
- Current address: The Animal Medical Center, New York, NY, USA
| | | | | | | | | | - Jessica M Quimby
- Jessica Quimby DVM, PhD, DACVIM (Internal Medicine), Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
47
|
Li Y, Li H, Cao Y, Wu F, Ma W, Wang Y, Sun S. Placenta‑derived mesenchymal stem cells improve airway hyperresponsiveness and inflammation in asthmatic rats by modulating the Th17/Treg balance. Mol Med Rep 2017; 16:8137-8145. [PMID: 28944907 PMCID: PMC5779899 DOI: 10.3892/mmr.2017.7605] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 08/04/2017] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess reparative and immunoregulatory properties, representing a hope for stem cell-based treatments. However, the mechanisms by which transplanted MSCs affect T helper (Th)17/regulatory T cell (Treg) balance in asthma patients remain unclear. The aim of the present study was to assess the therapeutic effects of human placenta MSCs (hPMSCs) in asthma, and explore the underlying mechanisms; in addition, the impact of hPMSCs transplantation on Th17/Treg balance in lymph and serum samples from asthmatic animals was evaluated. Sprague-Dawley rats were sensitized and challenged with ovalbumin (OVA). Administration of hPMSCs from human placenta resulted in increased Th17 and Treg in lymph samples compared with peripheral blood specimens. Enhanced pause values in OVA-treated animals were significantly higher than those in the control and hPMSCs treatment groups. The numbers of total cells, macrophages, neutrophils, and eosinophils were markedly increased in the OVA group compared with those of control + hPMSCs and control groups. In addition, interleukin 10, forkhead box P3 (Foxp3) and Treg levels in lymph, peripheral blood and lung tissue samples from asthma rats were increased significantly following hPMSC transplantation. Furthermore, Foxp3 protein levels increased, while those of RAR-related orphan receptor γ (RORγt) decreased after hPMSCs transplantation compared with the asthma group. Reduced IL-17, RORγt and Th17 levels were accompanied by reduced inflammatory cell infiltration, sub-epithelial smooth layer attenuation and mucus production in lung tissues. These results suggest that hPMSCs may improve airway hyperresponsiveness and inflammation by regulating the Th17/Treg balance in rats with asthma.
Collapse
Affiliation(s)
- Yingying Li
- Department of Pediatrics, Nephrology, Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hongbo Li
- Department of Respiratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yinyin Cao
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Fuling Wu
- Department of Pediatrics, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Wenbin Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yuesi Wang
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Shuzhen Sun
- Department of Pediatrics, Nephrology, Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
48
|
Chae HK, Song WJ, Ahn JO, Li Q, Lee BY, Kweon K, Park SC, Youn HY. Immunomodulatory effects of soluble factors secreted by feline adipose tissue-derived mesenchymal stem cells. Vet Immunol Immunopathol 2017; 191:22-29. [PMID: 28895862 DOI: 10.1016/j.vetimm.2017.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 07/28/2017] [Accepted: 07/30/2017] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have immunomodulatory functions and differentiation capacity, and their clinical use is increasing in veterinary species. Although MSCs have been applied in the treatment in various inflammatory diseases, mechanistic research on feline MSCs is lacking. Accordingly, in this study, we aimed to investigate the immunomodulatory mechanisms of MSCs isolated from feline adipose tissue (fATMSCs). fATMSCs from healthy cats were cultured in an appropriate manner and cocultured with transwell-separated allogeneic feline peripheral blood mononuclear cells (fPBMCs) and RAW264.7 murine macrophages. After 48h of coculture, RNA was extracted from RAW264.7 cells and fPBMCs. Cytokine expression in these cells was measured using quantitative real-time polymerase chain reaction (qRT-PCR) and compared according to the presence of fATMSCs. The mRNA levels of pro-inflammatory cytokines, e.g., tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase, and interleukin (IL)-1β, were significantly decreased in cocultures of mitogen-stimulated RAW264.7 cells with fATMSCs compared with that in the RAW264.7 cells control group. Additionally, changes in the expression of mRNAs extracted from fPBMCs were as follows: pro-inflammatory TNF-α, interferon-γ, and IL-6 were decreased, and anti-inflammatory IL-10 was increased during coculture of mitogen-stimulated allogeneic fPBMCs with fATMSCs. We also extracted RNA and collected supernatants from fATMSCs during transwell culture for measurement of the expression and secretion of soluble factors by qRT-PCR and enzyme-linked immunosorbent assays, respectively. The mRNA expression of immunomodulatory factors from fATMSCs, including cyclooxygenase-2 (COX-2), transforming growth factor (TGF)-β, indoleamine-2,3-dioxygenase (IDO) and hepatocyte growth factor, increased in the presence of RAW264.7 cells. Similarly, TGF-β, COX-2, and IDO mRNA expression and prostaglandin E2 (PGE2) secretion from fATMSCs increased in the presence of allogeneic fPBMCs. Finally, we measured the viability of fPBMCs under various conditions. Cell viability decreased in fPBMCs suspended in fATMSC-derived conditioned medium, and this reduction was alleviated in the group supplemented with NS-398 a PGE2 inhibitor. Our data suggested that soluble factors, including PGE2, secreted by fATMSCs played an important role in the immunomodulatory effects of these cells. These findings may be helpful in the application of fATMSCs to feline patients with immune-related diseases.
Collapse
Affiliation(s)
- Hyung-Kyu Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Woo-Jin Song
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Jin-Ok Ahn
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Qiang Li
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Bo-Yeon Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Kyeong Kweon
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Sang-Chul Park
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Korea.
| |
Collapse
|
49
|
Parys M, Kruger JM, Yuzbasiyan-Gurkan V. Evaluation of Immunomodulatory Properties of Feline Mesenchymal Stem Cells. Stem Cells Dev 2017; 26:776-785. [DOI: 10.1089/scd.2016.0041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Maciej Parys
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - John M. Kruger
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Vilma Yuzbasiyan-Gurkan
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| |
Collapse
|
50
|
Ahmadi M, Rahbarghazi R, Soltani S, Aslani MR, Keyhanmanesh R. Contributory Anti-Inflammatory Effects of Mesenchymal Stem Cells, Not Conditioned Media, On Ovalbumin-Induced Asthmatic Changes in Male Rats. Inflammation 2017; 39:1960-1971. [PMID: 27590236 DOI: 10.1007/s10753-016-0431-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Our aim in selecting an appropriate cell fraction and conditioned media (CM) was to achieve the suitable candidate for ameliorating long-term chronic asthmatic changes of respiratory tract. Thirty-six rats were classified into healthy and sensitized groups, which were further divided into three subgroups; rats received systemically 50 μl volume of PBS, CM, or 2 × 106 rat bone marrow-derived mesenchymal stem cells (rBMMSCs). Tracheal responsiveness (TR), immunologic responses, and recruitment of rBMMSCs into the lungs were evaluated. A high degree of TR and total WBC and percentages of eosinophils and neutrophils was significantly recorded in all sensitized groups rather than of controls (p < 0.001 to p < 0.05). Concurrently, a significant improvement of TR and eosinophil and neutrophil return toward normal levels was evident in sensitized rats receiving cells as compared to parallel asthmatic animals. Flow cytometric monitoring of lymphocyte subpopulation revealed a decrease in the number of CD3+CD4+ and concurrent increase in CD3+CD8+ in all sensitized rats as compared to control (p < 0.001 to p < 0.05). Noticeably, no significant modulatory effects of either cell or CM administration were achieved on the CD3+CD4+ and CD3+CD8+ populations in non-asthmatic rats. Corroborating our results, the number of CD3+CD4+ tended to increase (p < 0.05) which coincided with a decreased manner of CD3+CD8+ populations as compared to other asthmatic groups (p < 0.01 to p < 0.05). Moreover, stem cells could efficiently transmigrate to the lung parenchyma, albeit the dynamic of asthmatic changes stimulated the rate of recruited cells. Our study shed light on superior effects of mesenchymal stem cells, but not CM, in attenuating chronic asthmatic changes in the model of rat.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Soltani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Aslani
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|