1
|
Ahmadieh-Yazdi A, Karimi M, Afkhami E, Hajizadeh-Tafti F, Kuchakzadeh F, Yang P, Sheykhhasan M. Unveiling therapeutic potential: Adipose tissue-derived mesenchymal stem cells and their exosomes in the management of diabetes mellitus, wound healing, and chronic ulcers. Biochem Pharmacol 2024; 226:116399. [PMID: 38944396 DOI: 10.1016/j.bcp.2024.116399] [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: 01/31/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Diabetes mellitus (DM) is a pervasive global health issue with substantial morbidity and mortality, often resulting in secondary complications, including diabetic wounds (DWs). These wounds, arising from hyperglycemia, diabetic neuropathy, anemia, and ischemia, afflict approximately 15% of diabetic patients, with a considerable 25% at risk of lower limb amputations. The conventional approaches for chronic and diabetic wounds management involves utilizing various therapeutic substances and techniques, encompassing growth factors, skin substitutes and wound dressings. In parallel, emerging cell therapy approaches, notably involving adipose tissue-derived mesenchymal stem cells (ADMSCs), have demonstrated significant promise in addressing diabetes mellitus and its complications. ADMSCs play a pivotal role in wound repair, and their derived exosomes have garnered attention for their therapeutic potential. This review aimed to unravel the potential mechanisms and provide an updated overview of the role of ADMSCs and their exosomes in diabetes mellitus and its associated complications, with a specific focus on wound healing.
Collapse
Affiliation(s)
- Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Karimi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Afkhami
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Kuchakzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
2
|
Jeung SY, An JH, Kim SS, Youn HY. Safety of Gonadal Tissue-Derived Mesenchymal Stem Cell Therapy in Geriatric Dogs with Chronic Disease. Animals (Basel) 2024; 14:2134. [PMID: 39061596 PMCID: PMC11273526 DOI: 10.3390/ani14142134] [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: 05/12/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Ensuring the safety of mesenchymal stem cell (MSC) therapy is a fundamental requirement in clinical practice. This study aimed to assess the safety of using gonadal tissue-derived MSCs (n = 10) compared to the commonly utilized adipose tissue-derived MSCs (n = 9) in geriatric dogs with chronic diseases. All participants received allogeneic MSC therapy, and no allergic reactions due to allogeneic cell immunogenicity were noted. Both groups showed no adverse changes in physical exams or hematological parameters before and after therapy. Importantly, there were no instances of tumor formation or growth post-treatment in either group. The findings demonstrated that dogs treated with gonadal tissue-derived MSCs experienced no clinical adverse effects. However, clinical adverse effects were reported in one case of adipose tissue-derived MSC therapy. Despite limitations in monitoring beyond one year and constraints due to a small and diverse patient group, this pioneering study validates the safe use of gonadal tissue-derived MSCs in aged companion animals. It underscores the potential of utilizing tissues from neutering procedures to advance regenerative medicine and expand cell banks and therapy options for companion animals.
Collapse
Affiliation(s)
- So-Young Jeung
- VIP Animal Medical Center, Seoul 02830, Republic of Korea; (S.-Y.J.); (S.-S.K.)
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ju-Hyun An
- Laboratory of Veterinary Emergency and Critical Care, Department of Veterinary Clinical Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si 24341, Republic of Korea;
| | - Sung-Soo Kim
- VIP Animal Medical Center, Seoul 02830, Republic of Korea; (S.-Y.J.); (S.-S.K.)
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
3
|
Sharun K, Jambagi K, Kumar R, Gugjoo MB, Pawde AM, Tuli HS, Dhama K, Amarpal. Clinical applications of adipose-derived stromal vascular fraction in veterinary practice. Vet Q 2022; 42:151-166. [PMID: 35841195 PMCID: PMC9364732 DOI: 10.1080/01652176.2022.2102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Adipose tissue-derived stromal vascular fraction (AdSVF) comprises a heterogeneous cell population, including the multipotent mesenchymal stem cells, hematopoietic stem cells, immune cells, endothelial cells, fibroblasts, and pericytes. As such, multipotent adipose tissue-derived mesenchymal stem cells (AdMSCs), are one of the important components of AdSVF. Commonly used techniques to harvest AdSVF involve enzymatic or non-enzymatic methods. The enzymatic method is considered to be the gold standard technique due to its higher yield. The cellular components of AdSVF can be resuspended in normal saline, platelet-rich plasma, or phosphate-buffered saline to produce a ready-to-use solution. Freshly isolated AdSVF has exhibited promising osteogenic and vasculogenic capacity. AdSVF has already been proven to possess therapeutic potential for osteoarthritis management. It is also an attractive therapeutic option for enhancing wound healing. In addition, the combined use of AdSVF and platelet-rich plasma has an additive stimulatory effect in accelerating wound healing and can be considered an alternative to AdMSC treatment. It is also widely used for managing various orthopaedic conditions in clinical settings and has the potential for regenerating bone, cartilage, and tendons. Autologous AdSVF cells are used along with bone substitutes and other biological factors as an alternative to conventional bone grafting techniques owing to their promising osteogenic and vasculogenic capacity. It can also be used for treating osteonecrosis, meniscus tear, chondromalacia, and tendon injuries in veterinary practice. It has several advantages over in vitro expanded AdMSC, including precluding the need for culturing, reduced risk of cell contamination, and cost-effectiveness, making it ideal for clinical use.
Collapse
Affiliation(s)
- Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Kaveri Jambagi
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Rohit Kumar
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Mudasir Bashir Gugjoo
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Shuhama, Srinagar, Jammu and Kashmir-190006, India
| | - Abhijit M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Amarpal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| |
Collapse
|
4
|
Enhanced Repaired Enthesis Using Tenogenically Differentiated Adipose-Derived Stem Cells in a Murine Rotator Cuff Injury Model. Stem Cells Int 2022; 2022:1309684. [PMID: 35607399 PMCID: PMC9124132 DOI: 10.1155/2022/1309684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Rotator cuff tear (RCT) is among the most common shoulder injuries and is prone to rerupture after surgery. Selecting suitable subpopulations of stem cells as a new specific cell type of mesenchymal stem cells has been increasingly used as a potential therapeutic tool in regenerative medicine. In this study, murine adipose-derived SSEA-4+CD90+PDGFRA+ subpopulation cells were successfully sorted, extracted, and identified. These cells showed good proliferation and differentiation potential, especially in the direction of tendon differentiation, as evidenced by qRT-PCR and immunofluorescence. Subsequently, we established a murine rotator cuff injury model and repaired it with subpopulation cells. Our results showed that the subpopulation cells embedded in a fibrin sealant significantly improved the histological score, as well as the biomechanical strength of the repaired tendon enthesis at four weeks after surgery, compared with the other groups. Hence, these findings indicated that the subpopulation of cells could augment the repaired enthesis and lead to better outcomes, thereby reducing the retear rate after rotator cuff repair. Our study provides a potential therapeutic strategy for rotator cuff healing in the future.
Collapse
|
5
|
Di Bella S, Cannella V, Mira F, Di Marco P, Lastra A, Gucciardi F, Purpari G, Guercio A. The Effect of a 7 Year-Long Cryopreservation on Stemness Features of Canine Adipose-Derived Mesenchymal Stem Cells (cAD-MSC). Animals (Basel) 2021; 11:ani11061755. [PMID: 34208255 PMCID: PMC8230844 DOI: 10.3390/ani11061755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are used in therapy in animal models and veterinary medicine, due to their capacity of inducing tissue regeneration and immunomodulation. Their clinical application requires a ready off-the-shelf amount of viable therapeutics doses. For this purpose, it is useful to cryopreserve MSCs to gain a ready and controlled source of abundant autologous stem cells. We evaluated the effect of 7 years cryopreservation using 10% dimethyl sulfoxide (DMSO) with different fetal bovine serum (FBS) concentrations (from 10 to 90%) on different passages of MSCs isolated from canine adipose tissue (cAD-MSCs). The study aimed to evaluate the most adequate cell passage and FBS percentage for the long-term cryopreservation of cells by maintaining the stemness features. Phenotype morphology, cell viability, osteogenic and adipogenic differentiation potentials, proliferative potential and expression of pluripotency markers were analyzed in thawed cells and compared with fresh ones. We demonstrated that cells cryopreserved with at least 80% FBS maintain unaltered the stemness characteristics of the freshly isolated cells. In particular, cells of P0-P1 passages have to be expanded in vitro and subsequently cryopreserved and cells of P2-P4 passages should be considered in the studies on therapeutic application and in vitro study of cAD-MSCs.
Collapse
|
6
|
Satani N, Zhang X, Giridhar K, Wewior N, Cai C, Aronowski J, Savitz SI. A Combination of Atorvastatin and Aspirin Enhances the Pro-Regenerative Interactions of Marrow Stromal Cells and Stroke-Derived Monocytes In Vitro. Front Pharmacol 2021; 12:589418. [PMID: 33959001 PMCID: PMC8093790 DOI: 10.3389/fphar.2021.589418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/25/2021] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose: Marrow stromal cells (MSCs) are being tested in clinical trials for stroke patients. MSCs appear to promote recovery through secretomes that promote modulation of immune cells, including myeloid phagocytes. Many stroke patients have comorbidities such as metabolic syndrome, hypertension, hypercholesterolemia, and diabetes for which they are prescribed medications that might affect the function of MSCs and monocytes (Mo) when they are administered in stroke patients. We studied the effects of the two most commonly prescribed stroke medications, statin and statin plus aspirin, on the secretomes of MSCs and their modulation of Mo derived from stroke patients. Methods: Human MSCs, Mo and their co-cultures were exposed to atorvastatin or atorvastatin plus aspirin followed by secretome analysis at 24 h. Monocytes were isolated from healthy controls as well as stroke patients with NIHSS ranging from 11 to 20. Secretome composition was measured using multiplex immunoassay. We used MTT assay to measure proliferation of monocytes. The mixed model was used to analyze experimental data. p-values less than 0.05 were considered significant. Results: Atorvastatin and aspirin combination increased the release of IL-1RA from stroke Mo. In MSCs, atorvastatin and aspirin combination reduced the release of pro-inflammatory cytokines such as IL-6, IL-8, MCP-1 and IFN-γ. Atorvastatin alone reduced the release of IL-6, IL-8 and MCP-1 from co-cultures of stroke monocytes and MSCs. Combination of atorvastatin and aspirin had additive effect on reducing the secretion of IL-6 from co-cultures of stroke Mo and MSCs. Conclusion: Atorvastatin, alone and in combination with aspirin can promote anti-inflammatory effect by modulating the secretome profile of Mo and MSCs. Our results suggest that stroke trials involving the use of intravenous MSCs should consider the effect of aspirin and atorvastatin, both of which are administered to the majority of hospitalized ischemic stroke patients.
Collapse
Affiliation(s)
- Nikunj Satani
- Department of Neurology, Institute for Stroke and Cerebrovascular Disease, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Xu Zhang
- Center for Clinical and Translational Sciences, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Kaavya Giridhar
- Department of Neurology, Institute for Stroke and Cerebrovascular Disease, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Natalia Wewior
- Department of Neurology, Institute for Stroke and Cerebrovascular Disease, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Chunyan Cai
- Center for Clinical and Translational Sciences, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, Institute for Stroke and Cerebrovascular Disease, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Sean I Savitz
- Department of Neurology, Institute for Stroke and Cerebrovascular Disease, McGovern Medical School at UTHealth, Houston, TX, United States
| |
Collapse
|
7
|
Hendawy H, Uemura A, Ma D, Namiki R, Samir H, Ahmed MF, Elfadadny A, El-Husseiny HM, Chieh-Jen C, Tanaka R. Tissue Harvesting Site Effect on the Canine Adipose Stromal Vascular Fraction Quantity and Quality. Animals (Basel) 2021; 11:ani11020460. [PMID: 33572472 PMCID: PMC7916364 DOI: 10.3390/ani11020460] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 02/03/2023] Open
Abstract
Simple Summary Adipose stromal vascular fraction (SVF) cells are freshly isolated non-cultured mesenchymal stem cells, which have been recently applied in the treatment of several musculoskeletal inflammatory conditions in dogs. However, the best adipose tissue (AT) sampling site is still challenging. This study first addressed the ideal AT harvesting site in canines ranging between middle and old age, the most susceptible age to chronic musculoskeletal problems. Our results showed that the peri-ovarian region is the best AT harvesting site, which yields high amounts of SVF cells with enough adipose-derived stem cells. These data may help the further set-up of cell-based regenerative therapies at the preclinical and experimental level in canines. Abstract Mesenchymal stem cells (MSCs) constitute a great promise for regenerative therapy, but these cells are difficultly recovered in large amounts. A potent alternative is the stromal vascular fraction (SVF), non-cultured MSCs, separated from adipose tissue (AT). We aim to evaluate AT harvesting site effect on the SVF cells’ quantity and quality in dogs. Subcutaneous abdominal fat, falciform ligament and peri-ovarian fat were sampled. After SVF isolation, the trypan blue exclusion test and a hemocytometer were used to assess the cell viability and cellular yield. SVF cells were labeled for four surface antigenic markers, clusters of differentiation CD90, CD44, CD29, and CD45, and then examined by flow cytometry. Semi-quantitative RT-PCR was used to evaluate the gene expression of the former markers in addition to OCT-4 and CD34. SVF cells in the peri-ovarian AT recorded the highest viability% (99.63 ± 0.2%), as well as a significantly higher cellular yield (36.87 ± 19.6 × 106 viable cells/gm fat, p < 0.001) and a higher expression of adipose-derived mesenchymal stem cells AD-MSCs surface markers than that of other sites. SVF cells from the peri-ovarian site revealed a higher expression of MSC markers (CD90, CD44, and CD29) and OCT-4 compared to the other sites, with weak CD45 and CD34 expressions. The positive OCT-4 expression demonstrated the pluripotency of SVF cells isolated from different sites. To conclude, the harvesting site is a strong determinant of SVF cells’ quantity and quality, and the peri-ovarian site could be the best AT sampling site in dogs.
Collapse
Affiliation(s)
- Hanan Hendawy
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Akiko Uemura
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
| | - Danfu Ma
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
| | - Ryosuke Namiki
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
| | - Haney Samir
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Mahmoud F. Ahmed
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Ahmed Elfadadny
- Department of Animal Medicine, Faculty of Veterinary Medicine, Damanhur University, Damanhur, El-Beheira 22511, Egypt;
| | - Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Elqaliobiya 13736, Egypt
| | - Cheng Chieh-Jen
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (H.H.); (A.U.); (D.M.); (R.N.); (H.M.E.-H.); (C.C.-J.)
- Correspondence: ; Tel.: +81-042-367-5904
| |
Collapse
|
8
|
Voga M, Kovač V, Majdic G. Comparison of Canine and Feline Adipose-Derived Mesenchymal Stem Cells/Medicinal Signaling Cells With Regard to Cell Surface Marker Expression, Viability, Proliferation, and Differentiation Potential. Front Vet Sci 2021; 7:610240. [PMID: 33521084 PMCID: PMC7838367 DOI: 10.3389/fvets.2020.610240] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/26/2020] [Indexed: 12/23/2022] Open
Abstract
Remarkable immunomodulatory abilities of mesenchymal stem cells, also called multipotent mesenchymal stromal cells or medicinal signaling cells (MSCs), have entailed significant advances in veterinary regenerative medicine in recent years. Despite positive outcomes from MSC therapies in various diseases in dogs and cats, differences in MSC characteristics between small animal veterinary patients are not well-known. We performed a comparative study of cells' surface marker expression, viability, proliferation, and differentiation capacity of adipose-derived MSCs (ADMSCs) from dogs and domestic cats. The same growth media and methods were used to isolate, characterize, and culture canine and feline ADMSCs. Adipose tissue was collected from 11 dogs and 8 cats of both sexes. The expression of surface markers CD44, CD90, and CD34 was detected by flow cytometry. Viability at passage 3 was measured with the hemocytometer and compared to the viability measured by flow cytometry after 1 day of handling. The proliferation potential of MSCs was measured by calculating cell doubling and cell doubling time from second to eighth passage. Differentiation potential was determined at early and late passages by inducing cells toward adipogenic, osteogenic, and chondrogenic differentiation using commercial media. Our study shows that the percentage of CD44+CD90+ and CD34−/− cells is higher in cells from dogs than in cells from cats. The viability of cells measured by two different methods at passage 3 differed between the species, and finally, canine ADMSCs possess greater proliferation and differentiation potential in comparison to the feline ADMSCs.
Collapse
Affiliation(s)
- Metka Voga
- Veterinary Faculty, Institute for Preclinical Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Valerija Kovač
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Gregor Majdic
- Veterinary Faculty, Institute for Preclinical Sciences, University of Ljubljana, Ljubljana, Slovenia.,Medical Faculty, Institute for Physiology, University of Maribor, Maribor, Slovenia
| |
Collapse
|
9
|
Medications for Hypertension Change the Secretome Profile from Marrow Stromal Cells and Peripheral Blood Monocytes. Stem Cells Int 2020; 2020:8894168. [PMID: 32802081 PMCID: PMC7416264 DOI: 10.1155/2020/8894168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 07/15/2020] [Indexed: 01/01/2023] Open
Abstract
Marrow stromal cells (MSCs) are in different stages of clinical trials for stroke patients. MSCs are proposed to promote recovery through the release of secretomes that modulate the function of beneficial immune cells. The majority of stroke patients have comorbidities including hypertension, for which they are prescribed antihypertensive medications that might affect the function of MSCs, when they are administered in stroke patients. Here, we studied the effects of common antihypertensive medications on the secretomes of human MSCs and their modulation of human monocytes (Mo) derived from stroke patients. MTT assay was used to assess the proliferation of MSCs after they were exposed to increased levels of antihypertensive medications. MSCs were exposed to the following medications: atenolol, captopril, and losartan. Monocytes were isolated from stroke patients with NIHSS ranging from 11 to 20 and from healthy controls. MSC-Mo cocultures were established, and a secretome profile was analyzed using the Magpix Multiplex cytokine array from Luminex technology. The linear mixed-effect model was used for statistical analysis. All analyses were performed using SAS 9.4, and p values less than 0.05 were considered significant. At clinically relevant levels, there was no change in MSC proliferation after exposure to atenolol, captopril, or losartan. Atenolol increased IL-1RA in stroke-Mo and decreased IL-8 secretion from MSCs indicating an anti-inflammatory effect of atenolol on secretomes of these cells. Captopril increased IL-8 from stroke-Mo and increased IL-6, IL-8, and MCP-1 secretions from MSCs. Captopril also increased IL-6 secretion from cocultures of stroke-Mo and MSCs indicating a strong proinflammatory effect on MSCs and their interaction with Mo. Atenolol increased the secretion of IL-8 and MCP-1 while captopril increased the secretion of IL-6 and MCP-1 from MSCs. Losartan decreased the release of IL-6 from MSCs. Losartan reduced MCP-1 and TNF-α from stroke-Mo and reduced IL-8 from cocultures of stroke-Mo and MSCs. Our results show that antihypertensive medications such as atenolol, captopril, and losartan, at concentrations comparable to doses prescribed for patients hospitalized for acute stroke, modulate the secretome profile of MSCs and their modulatory effects on target immune cells. Our results suggest that stroke trials involving the use of intravenous MSCs should consider the effect of these antihypertensive drugs administered to stroke patients.
Collapse
|
10
|
Hu C, Zhao L, Li L. Current understanding of adipose-derived mesenchymal stem cell-based therapies in liver diseases. Stem Cell Res Ther 2019; 10:199. [PMID: 31287024 PMCID: PMC6613269 DOI: 10.1186/s13287-019-1310-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The liver, the largest organ with multiple synthetic and secretory functions in mammals, consists of hepatocytes, cholangiocytes, hepatic stellate cells (HSCs), sinusoidal endothelial cells, Kupffer cells (KCs), and immune cells, among others. Various causative factors, including viral infection, toxins, autoimmune defects, and genetic disorders, can impair liver function and result in chronic liver disease or acute liver failure. Mesenchymal stem cells (MSCs) from various tissues have emerged as a potential candidate for cell transplantation to promote liver regeneration. Adipose-derived MSCs (ADMSCs) with high multi-lineage potential and self-renewal capacity have attracted great attention as a promising means of liver regeneration. The abundance source and minimally invasive procedure required to obtain ADMSCs makes them superior to bone marrow-derived MSCs (BMMSCs). In this review, we comprehensively analyze landmark studies that address the isolation, proliferation, and hepatogenic differentiation of ADMSCs and summarize the therapeutic effects of ADMSCs in animal models of liver diseases. We also discuss key points related to improving the hepatic differentiation of ADMSCs via exposure of the cells to cytokines and growth factors (GFs), extracellular matrix (ECM), and various physical parameters in in vitro culture. The optimization of culturing methods and of the transplantation route will contribute to the further application of ADMSCs in liver regeneration and help improve the survival rate of patients with liver diseases. To this end, ADMSCs provide a potential strategy in the field of liver regeneration for treating acute or chronic liver injury, thus ensuring the availability of ADMSCs for research, trial, and clinical applications in various liver diseases in the future.
Collapse
Affiliation(s)
- Chenxia Hu
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| | - Lingfei Zhao
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| | - Lanjuan Li
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| |
Collapse
|
11
|
Aspirin in stroke patients modifies the immunomodulatory interactions of marrow stromal cells and monocytes. Brain Res 2019; 1720:146298. [PMID: 31220426 DOI: 10.1016/j.brainres.2019.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Most stroke patients are prescribed aspirin (ASA) to adjust blood coagulability. Marrow stromal cells (MSCs) are being tested in clinical trials for stroke patients who likely are prescribed aspirin. One of the principal mechanisms of action of MSCs and ASA is modulation of the inflammatory response, including those mediated by monocytes (Mo). Thus, here we tested if aspirin can modify anti-inflammatory properties of MSCs or Mo alone, and in combination. METHODS Mo were isolated at 24 h of stroke onset from ischemic stroke patients with NIHSS ranging from 11 to 20 or from healthy controls. Human bone marrow-derived MSCs from healthy subjects were used at passage 3. Mo, MSCs, and MSCs-Mo co-cultures were exposed to ASA at clinically relevant doses. The secretome profile of inflammatory mediators was measured using Magpix multiplex cytokine array. Viability was measured using MTT assay. Linear mixed effect model was used for statistical analysis. RESULTS Overall Mo from control subjects exposed to ASA showed increased secretion of IL-1RA, IL-8, MCP-1, and TNF-α and Mo from stroke patients showed greater release of IL-1RA and MCP-1. In MSCs-Mo co-cultures, ASA added to co-cultures of control Mo reduced fractalkine secretion while it increased the fractalkine secretion when added to Mo from stroke patients. In addition, in co-cultures independent of Mo origin, ASA reduced IL-6, IL-8, MCP-1, and TNF-α. CONCLUSIONS Aspirin in acute stroke patients may modulate the secretome profile of Mo and MSCs, thus potentially modulating immune and inflammatory responses associated with stroke. Our results suggest that stroke trials involving the use of intravenous MSCs should consider the effect of aspirin as a confounding factor.
Collapse
|
12
|
Oliva J, Florentino A, Bardag-Gorce F, Niihara Y. Engineering, differentiation and harvesting of human adipose-derived stem cell multilayer cell sheets. Regen Med 2019; 14:151-163. [PMID: 30829557 DOI: 10.2217/rme-2018-0053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Aim: The study goals were to engineer and harvest scaffold-free undifferentiated/differentiated multilayer human adipose-derived stem cell (hADSC) cell sheets, in absence of treatment. Materials & methods: The hADSC are seeded in 35 mm culture dishes. At confluence or when multilayer cell sheets are formed, hADSC are treated with predefined differentiation culture media (adipocyte, chondrocyte and osteoblast). Results: Undifferentiated hADSC and differentiated adipocyte, osteoblast and chondrocyte hADSC multilayer cell sheets (hADSCmCS) have been harvested. Hematoxylin & eosin showed the formation of multilayer cell sheets. Undifferentiated hADSC multilayer cell sheets preserve their stem cell markers. Differentiated adipocyte, osteoblast and chondrocyte hADSCmCS expressed specific markers. Conclusion: This simple protocol opens possibilities to engineer scaffold-free hADSCm cell sheet to transplant them on damaged organs.
Collapse
Affiliation(s)
- Joan Oliva
- Department of Medicine, LA BioMed at Harbor UCLA Medical Center, Torrance, CA 90502, USA.,Emmaus Life Sciences, Inc., 21250 Hawthorne Blvd., Suite 800, Torrance, CA 90503, USA
| | - Arjie Florentino
- Department of Medicine, LA BioMed at Harbor UCLA Medical Center, Torrance, CA 90502, USA
| | - Fawzia Bardag-Gorce
- Department of Medicine, LA BioMed at Harbor UCLA Medical Center, Torrance, CA 90502, USA
| | - Yutaka Niihara
- Department of Medicine, LA BioMed at Harbor UCLA Medical Center, Torrance, CA 90502, USA.,Emmaus Life Sciences, Inc., 21250 Hawthorne Blvd., Suite 800, Torrance, CA 90503, USA
| |
Collapse
|
13
|
Barboni B, Russo V, Berardinelli P, Mauro A, Valbonetti L, Sanyal H, Canciello A, Greco L, Muttini A, Gatta V, Stuppia L, Mattioli M. Placental Stem Cells from Domestic Animals: Translational Potential and Clinical Relevance. Cell Transplant 2019; 27:93-116. [PMID: 29562773 PMCID: PMC6434480 DOI: 10.1177/0963689717724797] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The field of regenerative medicine is moving toward clinical practice in veterinary science. In this context, placenta-derived stem cells isolated from domestic animals have covered a dual role, acting both as therapies for patients and as a valuable cell source for translational models. The biological properties of placenta-derived cells, comparable among mammals, make them attractive candidates for therapeutic approaches. In particular, stemness features, low immunogenicity, immunomodulatory activity, multilineage plasticity, and their successful capacity for long-term engraftment in different host tissues after autotransplantation, allo-transplantation, or xenotransplantation have been demonstrated. Their beneficial regenerative effects in domestic animals have been proven using preclinical studies as well as clinical trials starting to define the mechanisms involved. This is, in particular, for amniotic-derived cells that have been thoroughly studied to date. The regenerative role arises from a mutual tissue-specific cell differentiation and from the paracrine secretion of bioactive molecules that ultimately drive crucial repair processes in host tissues (e.g., anti-inflammatory, antifibrotic, angiogenic, and neurogenic factors). The knowledge acquired so far on the mechanisms of placenta-derived stem cells in animal models represent the proof of concept of their successful use in some therapeutic treatments such as for musculoskeletal disorders. In the next future, legislation in veterinary regenerative medicine will be a key element in order to certify those placenta-derived cell-based protocols that have already demonstrated their safety and efficacy using rigorous approaches and to improve the degree of standardization of cell-based treatments among veterinary clinicians.
Collapse
Affiliation(s)
- B Barboni
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Russo
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - P Berardinelli
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Mauro
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Valbonetti
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - H Sanyal
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Canciello
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Greco
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Muttini
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Gatta
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Stuppia
- 2 Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - M Mattioli
- 3 Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale," Teramo, Italy
| |
Collapse
|
14
|
Pereira MRDJ, Pinhatti VR, Silveira MDD, Matzenbacher CA, Freitas TROD, Silva JD, Camassola M, Nardi NB. Isolation and characterization of mesenchymal stem/stromal cells from Ctenomys minutus. Genet Mol Biol 2018; 41:870-877. [PMID: 30508008 PMCID: PMC6415614 DOI: 10.1590/1678-4685-gmb-2018-0012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/23/2018] [Indexed: 11/22/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells distributed in all tissues and characterized by adherence, morphology, immunophenotype and trilineage differentiation potential. The present study aimed to isolate and characterize adherent MSC-like populations from different tissues of Ctenomys minutus, a threatened wildlife rodent popularly known as tuco-tuco. Adherent cells were isolated from bone marrow, brain, liver, pancreas and adipose tissue of three adult animals collect in southern Brazil. Cultures showed typical morphology and proliferation potential. Adipose-derived MSCs showed trilineage potential. Cultures derived from adipose tissue, bone marrow and brain were immunophenotyped with negative results for CD31, CD44, CD45, CD106, and MHC class II, as well as strong positive results for CD29. Low fluorescence levels were seen for CD49d, CD90.2 and CD117. Cultures were negative for CD49e, except for brain-derived cultures that were weakly positive. CD11b was negative in adipose-derived MSCs, but positive in brain and bone marrow-derived cultures. The scratch assay showed high migration potential for pancreas and adipose tissue-derived cells. This study represents the first report of isolation and characterization of cultures having characteristics of MSCs from Ctenomys minutus. The collection of biological information for biobanks represents an important contribution to the creation of strategies for prevention of loss of genetic diversity.
Collapse
Affiliation(s)
| | | | | | | | | | - Juliana da Silva
- Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Melissa Camassola
- Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Nance Beyer Nardi
- Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil, Canoas, RS, Brazil
| |
Collapse
|
15
|
P2Y 2 and P2Y 6 receptor activation elicits intracellular calcium responses in human adipose-derived mesenchymal stromal cells. Purinergic Signal 2018; 14:371-384. [PMID: 30088129 PMCID: PMC6298923 DOI: 10.1007/s11302-018-9618-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue contains self-renewing multipotent cells termed mesenchymal stromal cells. In situ, these cells serve to expand adipose tissue by adipogenesis, but their multipotency has gained interest for use in tissue regeneration. Little is known regarding the repertoire of receptors expressed by adipose-derived mesenchymal stromal cells (AD-MSCs). The purpose of this study was to undertake a comprehensive analysis of purinergic receptor expression. Mesenchymal stromal cells were isolated from human subcutaneous adipose tissue and confirmed by flow cytometry. The expression profile of purinergic receptors was determined by quantitative real-time PCR and immunocytochemistry. The molecular basis for adenine and uracil nucleotide-evoked intracellular calcium responses was determined using Fura-2 measurements. All the known subtypes of P2X and P2Y receptors, excluding P2X2, P2X3 and P2Y12 receptors, were detected at the mRNA and protein level. ATP, ADP and UTP elicited concentration-dependent calcium responses in mesenchymal cells (N = 7–9 donors), with a potency ranking ADP (EC50 1.3 ± 1.0 μM) > ATP (EC50 2.2 ± 1.1 μM) = UTP (3.2 ± 2.8 μM). Cells were unresponsive to UDP (< 30 μM) and UDP-glucose (< 30 μM). ATP responses were attenuated by selective P2Y2 receptor antagonism (AR-C118925XX; IC50 1.1 ± 0.8 μM, 73.0 ± 8.5% max inhibition; N = 7 donors), and UTP responses were abolished. ADP responses were attenuated by the selective P2Y6 receptor antagonist, MRS2587 (IC50 437 ± 133nM, 81.0 ± 8.4% max inhibition; N = 6 donors). These data demonstrate that adenine and uracil nucleotides elicit intracellular calcium responses in human AD-MSCs with a predominant role for P2Y2 and P2Y6 receptor activation. This study furthers understanding about how human adipose-derived mesenchymal stromal cells can respond to external signalling cues.
Collapse
|
16
|
Yaneselli KM, Kuhl CP, Terraciano PB, de Oliveira FS, Pizzato SB, Pazza K, Magrisso AB, Torman V, Rial A, Moreno M, Llambí S, Cirne-Lima E, Maisonnave J. Comparison of the characteristics of canine adipose tissue-derived mesenchymal stem cells extracted from different sites and at different passage numbers. J Vet Sci 2018; 19:13-20. [PMID: 28693305 PMCID: PMC5799390 DOI: 10.4142/jvs.2018.19.1.13] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/25/2017] [Accepted: 05/05/2017] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have desirable characteristics for use in therapy in animal models and veterinary medicine, due to their capacity of inducing tissue regeneration and immunomodulation. The objective of this study was to evaluate the differences between canine adipose tissue-derived MSCs (AD-MSCs) extracted from subcutaneous (Sc) and visceral (Vs) sites. Surface antigenic markers, in vitro differentiation, and mineralized matrix quantification of AD-MSCs at different passages (P4, P6, and P8) were studied. Immunophenotypic analysis showed that AD-MSCs from both sites were CD44+, CD90+, and CD45-. Moreover, they were able, in vitro, to differentiate into fat, cartilage, and bone. Sc-AD-MSCs preserve in vitro multipotentiality up to P8, but Vs-AD-MSCs only tri-differentiated up to P4. In addition, compared to Vs-AD-MSCs, Sc-AD-MSCs had greater capacity for in vitro mineralized matrix synthesis. In conclusion, Sc-AD-MSCs have advantages over Vs-AD-MSCs, as Sc AD-MSCs preserve multipotentiality during a greater number of passages, have more osteogenic potential, and require less invasive extraction.
Collapse
Affiliation(s)
- Kevin M Yaneselli
- Laboratory of Immunology, Department of Microbiological Science, Faculty of Veterinary, Universidad de la República, Montevideo 11600, Uruguay
| | - Cristiana P Kuhl
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Paula B Terraciano
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Fernanda S de Oliveira
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Sabrina B Pizzato
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Kamila Pazza
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Alessandra B Magrisso
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Vanessa Torman
- Biostatistics, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Analía Rial
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Faculty of Medicine, Universidad de la República, Montevideo 11600, Uruguay
| | - María Moreno
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Faculty of Medicine, Universidad de la República, Montevideo 11600, Uruguay
| | - Silvia Llambí
- Laboratory of Genetics, Faculty of Veterinary, Universidad de la República, Montevideo 11600, Uruguay
| | - Elizabeth Cirne-Lima
- Laboratory of Embryology and Cellular Differentiation, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| | - Jacqueline Maisonnave
- Laboratory of Immunology, Department of Microbiological Science, Faculty of Veterinary, Universidad de la República, Montevideo 11600, Uruguay
| |
Collapse
|
17
|
Canine mesenchymal stem cells treated with TNF-α and IFN-γ enhance anti-inflammatory effects through the COX-2/PGE 2 pathway. Res Vet Sci 2018; 119:19-26. [PMID: 29783120 DOI: 10.1016/j.rvsc.2018.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/16/2018] [Accepted: 05/12/2018] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) have been used in studies on treatment of various diseases, and their application to immune-mediated diseases has garnered interest. Various methods for enhancing the immunomodulation effect of human MSCs have been used; however, similar approaches for canine MSCs are relatively unexplored. Accordingly, we evaluated immunomodulatory effects and mechanisms in canine MSCs treated with TNF-α and IFN-γ. Lipopolysaccharide (LPS)-stimulated RAW 264.7 cells were incubated with the conditioned media (CM) from canine MSCs for 48 h. Expression of RNA was assessed by quantitative reverse transcription PCR (qRT-PCR), and protein levels were assessed by western blot. Expression of inducible nitric oxide synthase (iNOS), IL-6 and IL-1β was significantly (one-way ANOVA) decreased in LPS-stimulated RAW 264.7 cells incubated with CM from canine MSCs compared to that in LPS-stimulated RAW 264.7 cells alone. Furthermore, anti-inflammatory effects of TNF-α- and IFN-γ-primed canine MSCs were significantly increased compared with those of naïve canine MSCs. Expression of cyclooxygenase 2 (COX-2) and prostaglandin E2 (PGE2) were likewise significantly increased in primed canine MSCs. The level of iNOS protein in LPS-stimulated RAW 264.7 cells incubated with CM from the primed canine MSCs was decreased, but it increased when the cells were treated with NS-398(PGE2 inhibitor). In conclusion, compared with naïve canine MSCs, cells primed with TNF-α and IFN-γ cause a greater reduction in release of anti-inflammatory cytokines from LPS-stimulated RAW 264.7 cells; the mechanism is upregulation of the COX-2/PGE2 pathway.
Collapse
|
18
|
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
|
19
|
Lee J, Lee KS, Kim CL, Byeon JS, Gu NY, Cho IS, Cha SH. Effect of donor age on the proliferation and multipotency of canine adipose-derived mesenchymal stem cells. J Vet Sci 2018; 18:141-148. [PMID: 27456768 PMCID: PMC5489460 DOI: 10.4142/jvs.2017.18.2.141] [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: 08/24/2015] [Revised: 04/19/2016] [Accepted: 06/08/2016] [Indexed: 01/08/2023] Open
Abstract
Research into adipose tissue-derived mesenchymal stem cells (AD-MSCs) has demonstrated the feasibility of their use in clinical applications due to their ease of isolation and abundance in adipose tissue. We isolated AD-MSCs from young and old dogs, and the cells were subjected to sequential sub-passaging from passage 1 (P1) to P7. Canine AD-MSCs (cAD-MSCs) were examined for proliferation kinetics, expression of molecules associated with self-renewal, expression of cell surface markers, and differentiation potentials at P3. Cumulative population doubling level was significantly higher in cAD-MSCs of young donors than in those of old donors. In addition, expressions of CD73, CD80, Oct3/4, Nanog, cell survival genes and differentiation potentials were significantly higher in young donors than in old donors. The present study suggests that donor age should be considered when developing cell-based therapies for clinical application of cAD-MSCs.
Collapse
Affiliation(s)
- Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Keum Sil Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Chan-Lan Kim
- 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
| | - 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
|
20
|
Bearden RN, Huggins SS, Cummings KJ, Smith R, Gregory CA, Saunders WB. In-vitro characterization of canine multipotent stromal cells isolated from synovium, bone marrow, and adipose tissue: a donor-matched comparative study. Stem Cell Res Ther 2017; 8:218. [PMID: 28974260 PMCID: PMC5627404 DOI: 10.1186/s13287-017-0639-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Background The dog represents an excellent large animal model for translational cell-based studies. Importantly, the properties of canine multipotent stromal cells (cMSCs) and the ideal tissue source for specific translational studies have yet to be established. The aim of this study was to characterize cMSCs derived from synovium, bone marrow, and adipose tissue using a donor-matched study design and a comprehensive series of in-vitro characterization, differentiation, and immunomodulation assays. Methods Canine MSCs were isolated from five dogs with cranial cruciate ligament rupture. All 15 cMSC preparations were evaluated using colony forming unit (CFU) assays, flow cytometry analysis, RT-PCR for pluripotency-associated genes, proliferation assays, trilineage differentiation assays, and immunomodulation assays. Data were reported as mean ± standard deviation and compared using repeated-measures analysis of variance and Tukey post-hoc test. Significance was established at p < 0.05. Results All tissue samples produced plastic adherent, spindle-shaped preparations of cMSCs. Cells were negative for CD34, CD45, and STRO-1 and positive for CD9, CD44, and CD90, whereas the degree to which cells were positive for CD105 was variable depending on tissue of origin. Cells were positive for the pluripotency-associated genes NANOG, OCT4, and SOX2. Accounting for donor and tissue sources, there were significant differences in CFU potential, rate of proliferation, trilineage differentiation, and immunomodulatory response. Synovium and marrow cMSCs exhibited superior early osteogenic activity, but when assessing late-stage osteogenesis no significant differences were detected. Interestingly, bone morphogenic protein-2 (BMP-2) supplementation was necessary for early-stage and late-stage osteogenic differentiation, a finding consistent with other canine studies. Additionally, synovium and adipose cMSCs proliferated more rapidly, displayed higher CFU potential, and formed larger aggregates in chondrogenic assays, although proteoglycan and collagen type II staining were subjectively decreased in adipose pellets as compared to synovial and marrow pellets. Lastly, cMSCs derived from all three tissue sources modulated murine macrophage TNF-α and IL-6 levels in a lipopolysaccharide-stimulated coculture assay. Conclusions While cMSCs from synovium, marrow, and adipose tissue share a number of similarities, important differences in proliferation and trilineage differentiation exist and should be considered when selecting cMSCs for translational studies. These results and associated methods will prove useful for future translational studies involving the canine model. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0639-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert N Bearden
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shannon S Huggins
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Kevin J Cummings
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Roger Smith
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, College of Medicine, Texas A&M University, College Station, TX, USA
| | - William B Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
21
|
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
|
22
|
Parsha K, Mir O, Satani N, Yang B, Guerrero W, Mei Z, Cai C, Chen PR, Gee A, Hanley PJ, Aronowski J, Savitz SI. Mesenchymal stromal cell secretomes are modulated by suspension time, delivery vehicle, passage through catheter, and exposure to adjuvants. Cytotherapy 2017; 19:36-46. [DOI: 10.1016/j.jcyt.2016.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 01/09/2023]
|
23
|
Vozzi G, Lucarini G, Dicarlo M, Andreoni C, Salvolini E, Ferretti C, Mattioli-Belmonte M. In vitro lifespan and senescent behaviour of human periosteal derived stem cells. Bone 2016; 88:1-12. [PMID: 27102545 DOI: 10.1016/j.bone.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/08/2016] [Accepted: 04/06/2016] [Indexed: 01/26/2023]
Abstract
Periosteum derived progenitor cells (PDPCs) represent promising mesenchymal stem cells (MSCs) for skeletal regeneration and to test bone cell based tissue engineering strategies. Most of regenerative medicine approaches based on MSCs require a noteworthy amount of cells that must be expanded in vitro prior to their use. As culture expansion method may impact on cell behaviour, we assessed the replicative and metabolic capacity (nitric oxide production and glucose consumption), senescence hallmarks of PDPCs serially passaged as well as the expression of selected genes specifically related to early osteoblastic differentiation, bone remodelling and stemness during PDPC sequential passaging. We also scouted a Systems Biology approach to examine and elucidate the experimental results through mathematical modelling and in silico simulations. PDPC subculture led to a progressive proliferative decline but, despite this, PDPCs maintained almost constant their metabolic activity. In vitro, senescent PDPCs displayed the typical "replicative senescence" features, involving p16 and not p53 in the regulation of this phenomenon. Gene expression analysis evidenced the tendency of sub-cultured PDPCs to increase the expression of genes involved in bone resorption. The mathematical analysis of the experimental results showed a strict similarity between replicative senescence and age-related changes, enabling the definition of an in silico model mimicking PDPC behaviour in terms of nitric oxide (NO) production. The relationship between NO production and subculture passages could represent a cutting edge "replicative senescence index". Overall, our findings suggest the possibility to use early-passage PDPCs for bone regenerative approaches based on the local recruitment of stem cells, whilst the later cell passages could be a suitable in vitro tool to validate scaffolds intended for bone regeneration in elderly subjects.
Collapse
Affiliation(s)
- Giovanni Vozzi
- Research Centre "E. Piaggio", Faculty of Engineering, University of Pisa, Largo Lucio Lazzarino, 2, 56126 Pisa, Italy
| | - Guendalina Lucarini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Manuela Dicarlo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Chiara Andreoni
- Research Centre "E. Piaggio", Faculty of Engineering, University of Pisa, Largo Lucio Lazzarino, 2, 56126 Pisa, Italy
| | - Eleonora Salvolini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Concetta Ferretti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy.
| |
Collapse
|
24
|
Markoski MM. Advances in the Use of Stem Cells in Veterinary Medicine: From Basic Research to Clinical Practice. SCIENTIFICA 2016; 2016:4516920. [PMID: 27379197 PMCID: PMC4917716 DOI: 10.1155/2016/4516920] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/30/2016] [Accepted: 05/16/2016] [Indexed: 06/01/2023]
Abstract
Today, several veterinary diseases may be treated with the administration of stem cells. This is possible because these cells present a high therapeutic potential and may be injected as autologous or allogenic, freshly isolated, or previously cultured. The literature supports that the process is safe and brings considerable benefits to animal health. Knowledge about how adult stem cells modulate the molecular signals to activate cell homing has also been increasingly determined, evidencing the mechanisms which enable cells to repair and regenerate injured tissues. Preclinical studies were designed for many animal models and they have contributed to the translation to the human clinic. This review shows the most commonly used stem cell types, with emphasis on mesenchymal stem cells and their mechanistic potential to repair, as well as the experimental protocols, studied diseases, and species with the highest amount of studies and applications. The relationship between stem cell protocols utilized on clinics, molecular mechanisms, and the physiological responses may offer subsidies to new studies and therefore improve the therapeutic outcome for both humans and animals.
Collapse
Affiliation(s)
- Melissa Medeiros Markoski
- Laboratório de Cardiologia Molecular e Celular, Fundação Universitária de Cardiologia/Instituto de Cardiologia, Princesa Isabel Avenue 370, 90620-001 Porto Alegre, RS, Brazil
| |
Collapse
|
25
|
Yang D, Wang ZQ, Deng JQ, Liao JY, Wang X, Xie J, Deng MM, Lü MH. Adipose-derived stem cells: A candidate for liver regeneration. J Dig Dis 2015; 16:489-98. [PMID: 26121206 DOI: 10.1111/1751-2980.12268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The scarcity of donor livers and the impracticality of hepatocyte transplantation represent the biggest obstacles for the treatment of liver failure. Adipose-derived stem cells, with their ability to differentiate into the hepatic lineage, provide a reliable alternative cell source with clear ethical and practical advantages. Moreover, adipose-derived stem cells can effectively repair liver damage by the dominant indirect pattern and increase the number of hepatocytes by the secondary direct pattern. In recent years, the development of the indirect pattern, which mainly includes immunomodulatory and trophic effects, has become a hot topic in the field of cell engineering. Therefore, adipose-derived stem cells are considered to be ideal therapeutic stem cells for human liver regeneration. In this article, we reviewed the advantages of adipose-derived stem cells in liver regeneration, and explore their underlying mechanisms.
Collapse
Affiliation(s)
- Dan Yang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Zhong Qiong Wang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Jia Qi Deng
- School of Foreign Languages of Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Jing Yuan Liao
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Xuan Wang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Jing Xie
- Department of Pediatric Surgery, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Ming Ming Deng
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Mu Han Lü
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| |
Collapse
|
26
|
Marx C, Silveira MD, Beyer Nardi N. Adipose-derived stem cells in veterinary medicine: characterization and therapeutic applications. Stem Cells Dev 2015; 24:803-13. [PMID: 25556829 DOI: 10.1089/scd.2014.0407] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells, considered one of the most promising cell types for therapeutic applications due to their capacity to secrete regenerative bioactive molecules, are present in all tissues. Stem cells derived from the adipose tissue have been increasingly used for cell therapy in humans and animals, both as freshly isolated, stromal vascular fraction (SVF) cells, or as cultivated adipose-derived stem cells (ASCs). ASCs have been characterized in different animal species for proliferation, differentiation potential, immunophenotype, gene expression, and potential for tissue engineering. Whereas canine and equine ASCs are well studied, feline cells are still poorly known. Many companies around the world offer ASC therapy for dogs, cats, and horses, although in most countries these activities are not yet controlled by regulatory agencies. This is the first study to review the characterization and clinical use of SVF and ASCs in spontaneously occurring diseases in veterinary patients. Although a relatively large number of studies investigating ASC therapy in induced lesions are available in the literature, a surprisingly small number of reports describe ASC therapy for naturally affected dogs, cats, and horses. A total of seven studies were found with dogs, only two studies in cats, and four in horses. Taken as a whole, the results do not allow a conclusion on the effect of this therapy, due to the generally small number of patients included, diversity of cell populations used, and lack of adequate controls. Further controlled studies are clearly needed to establish the real potential of ASC in veterinary medicine.
Collapse
Affiliation(s)
- Camila Marx
- 1 Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil , Canoas, RS, Brazil
| | | | | |
Collapse
|