1
|
Gu NY, Ryu GS, Park GN, Lee JY, Cho YS, Yang DK, Lee HJ. Enhanced susceptibility of porcine muscle-derived mesenchymal stem cells to Aujeszky's virus compared Vero cells. Anim Biotechnol 2025; 36:2479677. [PMID: 40110865 DOI: 10.1080/10495398.2025.2479677] [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: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Mesenchymal stem cells (MSCs) can self-renew and differentiate into several lineages and can be isolated from different tissues such as bone marrow, adipose tissue, umbilical cord blood, and muscle. Herein, we established MSCs derived from miniature pig muscle (MpMu-MSCs) and assessed their response to Aujeszky's virus. We characterized the MpMu-MSCs based on their cellular morphology, proliferation properties, cell surface marker expression, and mesodermal differentiation potential. MpMu-MSCs demonstrated a fibroblast-like spindle shape and formed a homogeneous monolayer. They showed a considerable increase in cell proliferation over 16 passages. The cells expressed surface markers CD29, CD44, CD90, and CD105 and demonstrated mesodermal lineage differentiation capabilities. MpMu-MSCs demonstrated faster cytopathic effects than the Vero cells when infected with Aujeszky's virus. The virus titer in MpMu-MSCs was initiated at 101.4 TCID50/ml at 12 h post-infection (hpi) and increased to 106.6 TCID50/ml at 72 hpi. In Vero cells, it was initiated at 102.3 TCID50/ml at 48 hpi and increased to 103.8 TCID50/ml at 72 hpi. This study showed that the stem cells procured from miniature pig muscles exhibit MSC characteristics and that the established cells demonstrate higher susceptibility and virus titer to Aujeszky's virus than Vero cells, indicating their potential use in virus research.
Collapse
Affiliation(s)
- Na-Yeon Gu
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Gwang Sik Ryu
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Gyu-Nam Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Ju-Yeon Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Yun Sang Cho
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Dong-Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Hye Jeong Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
2
|
Petinati N, Shipounova I, Sats N, Dorofeeva A, Sadovskaya A, Kapranov N, Tkachuk Y, Bondarenko A, Muravskaya M, Kotsky M, Kaplanskaya I, Vasilieva T, Drize N. Multipotent Mesenchymal Stromal Cells from Porcine Bone Marrow, Implanted under the Kidney Capsule, form an Ectopic Focus Containing Bone, Hematopoietic Stromal Microenvironment, and Muscles. Cells 2023; 12:268. [PMID: 36672203 PMCID: PMC9857022 DOI: 10.3390/cells12020268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are an object of intense investigation due to their therapeutic potential. MSCs have been well studied in vitro, while their fate after implantation in vivo has been poorly analyzed. We studied the properties of MSCs from the bone marrow (BM-MSC) before and after implantation under the renal capsule using a mini pig model. Autologous BM-MSCs were implanted under the kidney capsule. After 2.5 months, ectopic foci containing bones, foci of ectopic hematopoiesis, bone marrow stromal cells and muscle cells formed. Small pieces of the implant were cultivated as a whole. The cells that migrated out from these implants were cultured, cloned, analyzed and were proven to meet the most of criteria for MSCs, therefore, they are designated as MSCs from the implant-IM-MSCs. The IM-MSC population demonstrated high proliferative potential, similar to BM-MSCs. IM-MSC clones did not respond to adipogenic differentiation inductors: 33% of clones did not differentiate, and 67% differentiated toward an osteogenic lineage. The BM-MSCs revealed functional heterogeneity after implantation under the renal capsule. The BM-MSC population consists of mesenchymal precursor cells of various degrees of differentiation, including stem cells. These newly discovered properties of mini pig BM-MSCs reveal new possibilities in terms of their manipulation.
Collapse
Affiliation(s)
- Nataliya Petinati
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Irina Shipounova
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Natalia Sats
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Alena Dorofeeva
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Alexandra Sadovskaya
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
- Department of Immunology, Faculty of Biology, Federal State Budget Educational Institution of Higher Education M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nikolay Kapranov
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| | - Yulia Tkachuk
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Anatoliy Bondarenko
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Margarita Muravskaya
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Michail Kotsky
- Bioclinic for Working with Animals, Federal State Budgetary Scientific Institution Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Irina Kaplanskaya
- MNIOI Them. P.A. Herzen—Branch of the Federal State Budgetary Institution “NMITs Radiology” of the Ministry of Health of Russia, Department of Pathomorphology, 125284 Moscow, Russia
| | - Tamara Vasilieva
- Department of Cell Biology, Faculty of Biology, Federal State Budget Educational Institution of Higher Education M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nina Drize
- Laboratory for Physiology of Hematopoiesis, National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, 125167 Moscow, Russia
| |
Collapse
|
3
|
Uberti B, Plaza A, Henríquez C. Pre-conditioning Strategies for Mesenchymal Stromal/Stem Cells in Inflammatory Conditions of Livestock Species. Front Vet Sci 2022; 9:806069. [PMID: 35372550 PMCID: PMC8974404 DOI: 10.3389/fvets.2022.806069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) therapy has been a cornerstone of regenerative medicine in humans and animals since their identification in 1968. MSCs can interact and modulate the activity of practically all cellular components of the immune response, either through cell-cell contact or paracrine secretion of soluble mediators, which makes them an attractive alternative to conventional therapies for the treatment of chronic inflammatory and immune-mediated diseases. Many of the mechanisms described as necessary for MSCs to modulate the immune/inflammatory response appear to be dependent on the animal species and source. Although there is evidence demonstrating an in vitro immunomodulatory effect of MSCs, there are disparate results between the beneficial effect of MSCs in preclinical models and their actual use in clinical diseases. This discordance might be due to cells' limited survival or impaired function in the inflammatory environment after transplantation. This limited efficacy may be due to several factors, including the small amount of MSCs inoculated, MSC administration late in the course of the disease, low MSC survival rates in vivo, cryopreservation and thawing effects, and impaired MSC potency/biological activity. Multiple physical and chemical pre-conditioning strategies can enhance the survival rate and potency of MSCs; this paper focuses on hypoxic conditions, with inflammatory cytokines, or with different pattern recognition receptor ligands. These different pre-conditioning strategies can modify MSCs metabolism, gene expression, proliferation, and survivability after transplantation.
Collapse
Affiliation(s)
- Benjamin Uberti
- Instituto de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anita Plaza
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henríquez
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Claudio Henríquez
| |
Collapse
|
4
|
Sanooghi D, Amini N, Azedi F, Bagher Z, Parvishan A, Lotfi A, Rashidi N, Lotfi E, Sayahpour FA, Faghihi F. Differentiation of Mesenchymal Stem Cells Derived From Human Adipose Tissue Into Cholinergic-like Cells: An in Vitro Study. Basic Clin Neurosci 2021; 12:315-323. [PMID: 34917291 PMCID: PMC8666926 DOI: 10.32598/bcn.2021.1008.2] [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] [Received: 08/23/2019] [Revised: 10/10/2019] [Accepted: 02/15/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction: Cholinergic-associated diseases currently constitute a significant cause of neurological and neurodegenerative disabilities. As the drugs are not efficient in improving the suffered tissues, stem cell treatment is considered an effective strategy for substituting the lost cells. Methods: In the current study, we set out to investigate the differentiation properties of human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs) into cholinergic-like cells by two morphogens of Retinoic Acid (RA) and Sonic Hedgehog (Shh) using a three-step in vitro procedure. The results were evaluated using real-time PCR, flow cytometry, and immunocytochemistry for two weeks. Results: Our data showed that the cells could express cholinergic specific markers, including Islet-1, Acetylcholinesterase (AChE), SMI-32, and Nestin, at mRNA and protein levels. We could also quantitatively evaluate the expression of Islet-1, AChE, and Nestin at 14 days post-induction using flow cytometry. Conclusion: Human AD-MSCs are potent cells to differentiate into cholinergic-like cells in the presence of RA and Shh through a three-step protocol. Thus, they could be a suitable cell candidate for the regeneration of cholinergic-associated diseases. However, more functional and electrophysiological analyses are needed in this regard.
Collapse
Affiliation(s)
- Davood Sanooghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Center, The Five Basic Sensory Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicin, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asghar Parvishan
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Lotfi
- Damavand Agricultural College, Technical and Vocational University, Tehran, Iran
| | - Nooshin Rashidi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Lotfi
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Weatherall EL, Avilkina V, Cortes-Araya Y, Dan-Jumbo S, Stenhouse C, Donadeu FX, Esteves CL. Differentiation Potential of Mesenchymal Stem/Stromal Cells Is Altered by Intrauterine Growth Restriction. Front Vet Sci 2020; 7:558905. [PMID: 33251256 PMCID: PMC7676910 DOI: 10.3389/fvets.2020.558905] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Consistency in clinical outcomes is key to the success of therapeutic Mesenchymal Stem/Stromal cells (MSCs) in regenerative medicine. MSCs are used to treat both humans and companion animals (horses, dogs, and cats). The properties of MSC preparations can vary significantly with factors including tissue of origin, donor age or health status. We studied the effects of developmental programming associated with intrauterine growth restriction (IUGR) on MSC properties, particularly related to multipotency. IUGR results from inadequate uterine capacity and placental insufficiency of multifactorial origin. Both companion animals (horses, dogs, cats) and livestock (pigs, sheep, cattle) can be affected by IUGR resulting in decreased body size and other associated changes that can include, alterations in musculoskeletal development and composition, and increased adiposity. Therefore, we hypothesized that this dysregulation occurs at the level of MSCs, with the cells from IUGR animals being more prone to differentiate into adipocytes and less to other lineages such as chondrocytes and osteocytes compared to those obtained from normal animals. IUGR has consequences on health and performance in adult life and in the case of farm animals, on meat quality. In humans, IUGR is linked to increased risk of metabolic (type 2 diabetes) and other diseases (cardiovascular), later in life. Here, we studied porcine MSCs where IUGR occurs spontaneously, and shows features that recapitulate human IUGR. We compared the properties of adipose-derived MSCs from IUGR (IUGR-MSCs) and Normal (Normal-MSCs) new-born pig littermates. Both MSC types grew clonally and expressed typical MSC markers (CD105, CD90, CD44) at similar levels. Importantly, tri-lineage differentiation capacity was significantly altered by IUGR. IUGR-MSCs had higher adipogenic capacity than Normal-MSCs as evidenced by higher adipocyte content and expression of the adipogenic transcripts, PPARγ and FABP4 (P < 0.05). A similar trend was observed for fibrogenesis, where, upon differentiation, IUGR-MSCs expressed significantly higher levels of COL1A1 (P < 0.03) than Normal-MSCs. In contrast, chondrogenic and osteogenic potential were decreased in IUGR-MSCs as shown by a smaller chondrocyte pellet and osteocyte staining, and lower expression of SOX9 (P < 0.05) and RUNX2 (P < 0.02), respectively. In conclusion, the regenerative potential of MSCs appears to be determined prenatally in IUGR and this should be taken into account when selecting cell donors in regenerative therapy programmes both in humans and companion animals.
Collapse
Affiliation(s)
- Emma L Weatherall
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Viktorija Avilkina
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Yennifer Cortes-Araya
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Susan Dan-Jumbo
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Claire Stenhouse
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| | - Francesc X Donadeu
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom.,The Euan Macdonald Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Cristina L Esteves
- The Roslin Institute and The Royal (DICK) School of Veterinary Studies (R(D)SVS), The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Yang Z, Vajta G, Xu Y, Luan J, Lin M, Liu C, Tian J, Dou H, Li Y, Liu T, Zhang Y, Li L, Yang W, Bolund L, Yang H, Du Y. Production of Pigs by Hand-Made Cloning Using Mesenchymal Stem Cells and Fibroblasts. Cell Reprogram 2017; 18:256-63. [PMID: 27459584 DOI: 10.1089/cell.2015.0072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) exhibited self-renewal and less differentiation, making the MSCs promising candidates for adult somatic cell nuclear transfer (SCNT). In this article, we tried to produce genome identical pigs through hand-made cloning (HMC), with MSCs and adult skin fibroblasts as donor cells. MSCs were derived from either adipose tissue or peripheral blood (aMSCs and bMSCs, respectively). MSCs usually showed the expression pattern of CD29, CD73, CD90, and CD105 together with lack of expression of the hematopoietic markers CD34and CD45. Flow cytometry results demonstrated high expression of CD29 and CD90 in both MSC lines, while CD73, CD34, and CD45 expression were not detected. In contrary, in reverse transcription-polymerase chain reaction (RT-PCR) analysis, CD73 and CD34 were detected indicating that human antibodies CD73 and CD34 were not suitable to identify porcine cell surface markers and porcine MSC cellular surface markers of CD34 might be different from other species. MSCs also had potential to differentiate successfully into chondrocytes, osteoblasts, and adipocytes. After HMC, embryos reconstructed with aMSCs had higher blastocyst rate on day 5 and 6 than those reconstructed with bMSCs and fibroblasts (29.6% ± 1.3% and 41.1% ± 1.4% for aMSCs vs. 23.9% ± 1.2% and 35.5% ± 1.6% for bMSCs and 22.1% ± 0.9% and 33.3% ± 1.1% for fibroblasts, respectively). Live birth rate per transferred blastocyst achieved with bMSCs (1.59%) was the highest among the three groups. This article was the first report to compare the efficiency among bMSCs, aMSCs, and fibroblasts for boar cloning, which offered a realistic perspective to use the HMC technology for commercial breeding.
Collapse
Affiliation(s)
- Zhenzhen Yang
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| | - Gábor Vajta
- 2 BGI-Shenzhen , Shenzhen, China .,3 Central Queensland University , Rockhampton, Australia
| | - Ying Xu
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| | - Jing Luan
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| | - Mufei Lin
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| | - Cong Liu
- 2 BGI-Shenzhen , Shenzhen, China
| | - Jianing Tian
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Hongwei Dou
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Yong Li
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| | - Tianbin Liu
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Yijie Zhang
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Lin Li
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Wenxian Yang
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China
| | - Lars Bolund
- 2 BGI-Shenzhen , Shenzhen, China .,4 Department of Biomedicine, University of Aarhus , Aarhus C, Denmark
| | | | - Yutao Du
- 1 BGI Ark Biotechnology Co., LTD (BAB) , Shenzhen, China .,2 BGI-Shenzhen , Shenzhen, China
| |
Collapse
|
7
|
Liu Z, Hu W, He T, Dai Y, Hara H, Bottino R, Cooper DKC, Cai Z, Mou L. Pig-to-Primate Islet Xenotransplantation: Past, Present, and Future. Cell Transplant 2017; 26:925-947. [PMID: 28155815 PMCID: PMC5657750 DOI: 10.3727/096368917x694859] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 03/21/2017] [Indexed: 12/17/2022] Open
Abstract
Islet allotransplantation results in increasing success in treating type 1 diabetes, but the shortage of deceased human donor pancreata limits progress. Islet xenotransplantation, using pigs as a source of islets, is a promising approach to overcome this limitation. The greatest obstacle is the primate immune/inflammatory response to the porcine (pig) islets, which may take the form of rapid early graft rejection (the instant blood-mediated inflammatory reaction) or T-cell-mediated rejection. These problems are being resolved by the genetic engineering of the source pigs combined with improved immunosuppressive therapy. The results of pig-to-diabetic nonhuman primate islet xenotransplantation are steadily improving, with insulin independence being achieved for periods >1 year. An alternative approach is to isolate islets within a micro- or macroencapsulation device aimed at protecting them from the human recipient's immune response. Clinical trials using this approach are currently underway. This review focuses on the major aspects of pig-to-primate islet xenotransplantation and its potential for treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhengzhao Liu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Wenbao Hu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Tian He
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Hidetaka Hara
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, USA
| | - David K. C. Cooper
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| |
Collapse
|
8
|
Almalki SG, Agrawal DK. ERK signaling is required for VEGF-A/VEGFR2-induced differentiation of porcine adipose-derived mesenchymal stem cells into endothelial cells. Stem Cell Res Ther 2017; 8:113. [PMID: 28499402 PMCID: PMC5429549 DOI: 10.1186/s13287-017-0568-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 12/26/2022] Open
Abstract
Background Cell-based therapy that can rejuvenate the endothelium with stimulated adipose-derived mesenchymal stem cells (AMSCs) is a promising therapeutic strategy for the re-endothelialization of denuded arteries at the stenting site. Previously, we have shown that silencing of MMP-2 and MMP-14 inhibits vascular endothelial growth factor receptor type 2 (VEGFR2) cleavage, and induces differentiation of AMSCs toward the endothelial cell (EC) lineage. In this study, we examined the underlying signaling pathways that regulate differentiation of AMSCs to ECs in vitro through VEGFR2. Methods AMSCs were isolated from porcine abdominal adipose tissue. The isolated AMSCs were characterized by positive expression of CD29, CD44, and CD90 and negative expression of CD11b and CD45. The isolated MSCs were transfected with siRNA to silence MMP-2, MMP-14, and angiotensin receptor 2 (ATR2). Cells were suspended either in endothelial basal media (EBM) or endothelial growth media (EGM) with various treatments. Flow cytometry was performed to examine the expression of EC markers, and western blot analysis was performed to examine the expression and activity of various kinases. Scratch assay was performed to examine the cell migration. Data were analyzed by ANOVA using PRISM GraphPad. Results After 10 days of stimulation for EC differentiation, the morphology of AMSCs changed to a morphology similar to that of ECs. Silencing MMP-2 and MMP-14 resulted in significant decrease in the number of migrated cells compared with the EGM-only group. ATR2 siRNA transfection did not affect the migration and differentiation of AMSCs to ECs. Stimulation of AMSCs for EC differentiation with or without MMP-2 or MMP-14 siRNA resulted in significant increase in p-ERK, and significant decrease in p-JNK. There was no significant change in p-p38 in all three groups compared with the EBM group. ERK inhibition resulted in significant decrease in the expression of EC markers in the EGM, EGM + MMP-2 siRNA, and EGM + MMP-14 siRNA groups. The VEGFR2 kinase inhibitor induced a dose-dependent inhibition of ERK. Conclusion The ERK signaling pathway is critical for VEGF-A/VEGFR2-induced differentiation of AMSCs into ECs. These findings provide new insights into the role of the ERK signaling pathway in AMSC differentiation to ECs for potential clinical use in cardiovascular diseases.
Collapse
Affiliation(s)
- Sami G Almalki
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, 68178, USA.
| |
Collapse
|
9
|
Almalki SG, Llamas Valle Y, Agrawal DK. MMP-2 and MMP-14 Silencing Inhibits VEGFR2 Cleavage and Induces the Differentiation of Porcine Adipose-Derived Mesenchymal Stem Cells to Endothelial Cells. Stem Cells Transl Med 2017; 6:1385-1398. [PMID: 28213979 PMCID: PMC5442711 DOI: 10.1002/sctm.16-0329] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 12/27/2022] Open
Abstract
The molecular mechanisms that control the ability of adipose‐derived mesenchymal stem cells (AMSCs) to remodel three‐dimensional extracellular matrix barriers during differentiation are not clearly understood. Herein, we studied the expression of matrix metalloproteinases (MMPs) during the differentiation of AMSCs to endothelial cells (ECs) in vitro. MSCs were isolated from porcine abdominal adipose tissue, and characterized by immunopositivity to CD44, CD90, CD105, and immunonegativity to CD14 and CD45. Plasticity of AMSCs was confirmed by multilineage differentiation. The mRNA transcripts for MMPs and Tissue Inhibitor of Metalloproteinases (TIMPs), and protein expression of EC markers were analyzed. The enzyme activity and protein expression were analyzed by gelatin zymography, enzyme‐linked immunosorbent assay (ELISA), and Western blot. The differentiation of AMSCs to ECs was confirmed by mRNA and protein expressions of the endothelial markers. The mRNA transcripts for MMP‐2 and MMP‐14 were significantly increased during the differentiation of MSCs into ECs. Findings revealed an elevated MMP‐14 and MMP‐2 expression, and MMP2 enzyme activity. Silencing of MMP‐2 and MMP‐14 significantly increased the expression of EC markers, formation of capillary tubes, and acetylated‐low‐density lipoprotein uptake, and decreased the cleavage of vascular endothelial growth factor receptor type 2 (VEGFR2). Inhibition of VEGFR2 significantly decreased the expression of EC markers. These novel findings demonstrate that the upregulation of MMP2 and MMP14 has an inhibitory effect on the differentiation of AMSCs to ECs, and silencing these MMPs inhibit the cleavage of VEGFR2 and stimulate the differentiation of AMSCs to ECs. These findings provide a potential mechanism for the regulatory role of MMP‐2 and MMP‐14 in the re‐endothelialization of coronary arteries following intervention. Stem Cells Translational Medicine2017;6:1385–1398
Collapse
Affiliation(s)
- Sami G Almalki
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Yovani Llamas Valle
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| |
Collapse
|
10
|
Valle YL, Almalki SG, Agrawal DK. Vitamin D machinery and metabolism in porcine adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2016; 7:118. [PMID: 27530414 PMCID: PMC4988022 DOI: 10.1186/s13287-016-0382-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/09/2016] [Accepted: 07/29/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Vitamin D, a hormone once thought to have a role limited to calcium homeostasis and bone mineralization, has pleiotropic effects on different types of cells. Vitamin D receptors are reported in vascular smooth muscle cells, endothelial cells, and cardiomyocytes. Adipose-derived MSCs (ADMSCs) are multipotent cells with the capacity to differentiate into cells of different lineages. To our knowledge, the presence of vitamin D machinery on porcine ADMSCs has not yet been examined. In this study, we investigated the presence of vitamin D machinery and metabolism in ADMSCs by analyzing the expression levels of vitamin D receptor (VDR), vitamin D metabolizing enzymes (CYP24A1 and CYP27B1) after in vitro stimulation with active vitamin D, calcitriol. METHODS AND RESULTS ADMSCs isolated from porcine adipose tissue were characterized by positive staining for ADMSC markers, CD44, CD73, and CD90, and negative staining for macrophage marker CD11b and hematopoietic stem cell markers CD34 and CD45, and trilineage differentiation to osteocytes, chondrocytes, and adipocytes. No cytotoxicity was observed when MSCs were stimulated with 0.1-10 nM calcitriol. The ADMSCs were analyzed for mRNA and protein expression of CYP24A1, CYP27B1, and VDR by immunostaining, qPCR, and ELISA. A significant increase (p <0.01) in the mRNA expression of CYP24A1, CYP27B1, and VDR was observed after stimulation of ADMSCs with calcitriol (10 nM). The in vitro time-dependent effect of calcitriol (10 nM) on the components of vitamin D machinery in cultured MSCs was determined by qPCR. The VDR and CYP27B1 expression peaked at 3 h and CYP24A1 at 24 h, respectively. The in vitro biosynthesis of 1, 25(OH)2D3 by ADMSCs was analyzed by ELISA and Western blot. The levels of the active form of vitamin D were significantly decreased once the CYP enzymes were inhibited (p <0.01), demonstrating the ability of ADMSCs to convert inactive vitamin D into active vitamin D for cellular action. CONCLUSIONS Porcine ADMSCs possess vitamin D hydrolases and VDR to metabolize and respond to vitamin D. Hence, in vivo circulating 25-hydroxy vitamin D levels may have a significant role in regulating the differentiation of ADMSCs into different lineages, which might assist in stem cell-based therapy.
Collapse
Affiliation(s)
- Yovani Llamas Valle
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Sami G. Almalki
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178 USA
| |
Collapse
|
11
|
Cooper DKC, Ezzelarab MB, Hara H, Iwase H, Lee W, Wijkstrom M, Bottino R. The pathobiology of pig-to-primate xenotransplantation: a historical review. Xenotransplantation 2016; 23:83-105. [PMID: 26813438 DOI: 10.1111/xen.12219] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/22/2015] [Indexed: 12/16/2022]
Abstract
The immunologic barriers to successful xenotransplantation are related to the presence of natural anti-pig antibodies in humans and non-human primates that bind to antigens expressed on the transplanted pig organ (the most important of which is galactose-α1,3-galactose [Gal]), and activate the complement cascade, which results in rapid destruction of the graft, a process known as hyperacute rejection. High levels of elicited anti-pig IgG may develop if the adaptive immune response is not prevented by adequate immunosuppressive therapy, resulting in activation and injury of the vascular endothelium. The transplantation of organs and cells from pigs that do not express the important Gal antigen (α1,3-galactosyltransferase gene-knockout [GTKO] pigs) and express one or more human complement-regulatory proteins (hCRP, e.g., CD46, CD55), when combined with an effective costimulation blockade-based immunosuppressive regimen, prevents early antibody-mediated and cellular rejection. However, low levels of anti-non-Gal antibody and innate immune cells and/or platelets may initiate the development of a thrombotic microangiopathy in the graft that may be associated with a consumptive coagulopathy in the recipient. This pathogenic process is accentuated by the dysregulation of the coagulation-anticoagulation systems between pigs and primates. The expression in GTKO/hCRP pigs of a human coagulation-regulatory protein, for example, thrombomodulin, is increasingly being associated with prolonged pig graft survival in non-human primates. Initial clinical trials of islet and corneal xenotransplantation are already underway, and trials of pig kidney or heart transplantation are anticipated within the next few years.
Collapse
Affiliation(s)
- David K C Cooper
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohamed B Ezzelarab
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hidetaka Hara
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hayato Iwase
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Whayoung Lee
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Wijkstrom
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Lee J, Byeon JS, Lee KS, Gu NY, Lee GB, Kim HR, Cho IS, Cha SH. Chondrogenic potential and anti-senescence effect of hypoxia on canine adipose mesenchymal stem cells. Vet Res Commun 2015; 40:1-10. [PMID: 26661466 DOI: 10.1007/s11259-015-9647-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/19/2015] [Indexed: 12/24/2022]
Abstract
Mesenchymal stem cells (MSCs) have the ability to differentiate into multi-lineage cells, which confers great promise for use in regenerative medicine. In this study, canine adipose MSCs (cAD-MSCs) were isolated from canine adipose tissue. These cells clearly represented stemness (Oct4, Sox2, and Nanog) and differentiation potential into the mesoderm (adipocytes, chondrocytes, and osteoblasts) at early passages. The aim of this study was to evaluate the effects of hypoxia on the differentiation potential into mesoderm, and the expression of anti-apoptotic genes associated with cell survival for the optimal culturing of MSCs. We observed that the proliferation of the cAD-MSCs meaningfully increased when cultured under hypoxic condition than in normoxic condition, during 7 consecutive passages. Also, we found that hypoxia strongly expressed anti-senescence related genes such as HDAC1 (histone deacetylase 1), DNMT1 (DNA (cytosine-5)-methyltransferase 1), Bcl-2 (inhibitor of apoptosis), TERT (telomerase reverse transcriptase), LDHA (lactate dehydrogenase A), SLC2A1 (glucose transporter), and DKC1 (telomere holoenzyme complex) and differentiation potential of cAD-MSCs into chondrocytes, than seen under the normoxic culture conditions. We also examined the multipotency of hypoxic conditioned MSCs using quantitative real-time RT-PCR. We found that the expression levels of stemness genes such as Oct-4, Nanog, and Sox-2 were increased in hypoxic condition when compared to the normoxic condition. Collectively, these results suggest that hypoxic conditions have the ability to induce proliferation of MSCs and augment their chondrogenic potential. This study suggests that cell proliferation of cAD-MSC under hypoxia could be beneficial, when considering these cells for cell therapies of canine bone diseases.
Collapse
Affiliation(s)
- Jienny Lee
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Jeong Su Byeon
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Keum Sil Lee
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Na-Yeon Gu
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Gyeong Been Lee
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Hee-Ryang Kim
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - In-Soo Cho
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea
| | - Sang-Ho Cha
- Animal Stem Cells Laboratory, Viral Disease Division, Animal and Plant Quarantine Agency, 175 Anyang-ro, Manan-gu, Anyang-si, Gyeonggi-do, 14089, Republic of Korea.
| |
Collapse
|
13
|
Higginbotham L, Ford ML, Newell KA, Adams AB. Preventing T cell rejection of pig xenografts. Int J Surg 2015; 23:285-290. [PMID: 26306770 DOI: 10.1016/j.ijsu.2015.07.722] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/30/2015] [Indexed: 11/25/2022]
Abstract
Xenotransplantation is a potential solution to the limited supply of donor organs. While early barriers to xenograft acceptance, such as hyperacute rejection, are now largely avoided through genetic engineering, the next frontier in successful xenograft survival will require prevention of T cell-mediated rejection. Most successful immunosuppressive regimens in xenotransplantation utilize T cell depletion with antibody therapy. Additionally, the use of T cell costimulatory blockade - specifically blockade of the CD40-CD154 pathway - shows promise with several reports of long-term xenograft survival. Additional therapies, such as transgenic expression of T cell coinhibitory molecules or transfer of immunomodulatory cells to promote tolerance, may be necessary to achieve reliable long-term xenograft acceptance. Further studies in pre-clinical models are essential in order to optimize these regimens prior to trials in patients.
Collapse
Affiliation(s)
- Laura Higginbotham
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mandy L Ford
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kenneth A Newell
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew B Adams
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Comparative studies on proliferation, molecular markers and differentiation potential of mesenchymal stem cells from various tissues (adipose, bone marrow, ear skin, abdominal skin, and lung) and maintenance of multipotency during serial passages in miniature pig. Res Vet Sci 2015; 100:115-24. [DOI: 10.1016/j.rvsc.2015.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 02/11/2015] [Accepted: 03/01/2015] [Indexed: 12/16/2022]
|
15
|
The potential role of genetically-modified pig mesenchymal stromal cells in xenotransplantation. Stem Cell Rev Rep 2015; 10:79-85. [PMID: 24142483 DOI: 10.1007/s12015-013-9478-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mesenchymal stromal cells (MSCs) are known to have regenerative, anti-inflammatory, and immunodulatory effects. There are extensive indications that pig MSCs function satisfactorily across species barriers. Pig MSCs might have considerable therapeutic potential, particularly in xenotransplantation, where they have several potential advantages. (i) pMSCs can be obtained from the specific organ- or cell-source donor pig or from an identical (cloned) pig. (ii) They are easy to obtain in large numbers, negating the need for prolonged ex vivo expansion. (iii) They can be obtained from genetically-engineered pigs, and the genetic modification can be related to the therapeutic goal of the MSCs. We have reviewed our own studies on MSCs from genetically-engineered pigs, and summarize them here. We have successfully harvested and cultured MSCs from wild-type and genetically-engineered pig bone marrow and adipose tissue. We have identified several pig (p)MSC surface markers (positive for CD29, CD44, CD73, CD105, CD166, and negative for CD31, CD45), have demonstrated their proliferation and differentiation (into adipocytes, osteoblasts, and chondroblasts), and evaluated their antigenicity and immune suppressive effects on human peripheral blood mononuclear cells and CD4(+)T cells. They have identical or very similar characteristics to MSCs from other mammals. Genetically-modified pMSCs are significantly less immunogenic than wild-type pMSCs, and downregulate the human T cell response to pig antigens as efficiently as do human MSCs. We hypothesized that pMSCs can immunomodulate human T cells through induction of apoptosis or anergy, or cause T cell phenotype switching with induction of regulatory T cells, but we could find no evidence for these mechanisms. However, pMSCs upregulated the expression of CD69 on human CD4(+) and CD8(+) T cells, the relevance of which is currently under investigation. We conclude that MSCs from genetically-engineered pigs should continue to be investigated for their immunomodulatory (and regenerative and anti-inflammatory) effects in pig-to-nonhuman primate organ and cell transplantation models.
Collapse
|
16
|
Allen AB, Priddy LB, Li MTA, Guldberg RE. Functional augmentation of naturally-derived materials for tissue regeneration. Ann Biomed Eng 2014; 43:555-67. [PMID: 25422160 DOI: 10.1007/s10439-014-1192-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/13/2014] [Indexed: 12/12/2022]
Abstract
Tissue engineering strategies have utilized a wide spectrum of synthetic and naturally-derived scaffold materials. Synthetic scaffolds are better defined and offer the ability to precisely and reproducibly control their properties, while naturally-derived scaffolds typically have inherent biological and structural properties that may facilitate tissue growth and remodeling. More recently, efforts to design optimized biomaterial scaffolds have blurred the line between these two approaches. Naturally-derived scaffolds can be engineered through the manipulation of intrinsic properties of the pre-existing backbone (e.g., structural properties), as well as the addition of controllable functional components (e.g., biological properties). Chemical and physical processing techniques used to modify structural properties of synthetic scaffolds have been tailored and applied to naturally-derived materials. Such strategies include manipulation of mechanical properties, degradation, and porosity. Furthermore, biofunctional augmentation of natural scaffolds via incorporation of exogenous cells, proteins, peptides, or genes has been shown to enhance functional regeneration over endogenous response to the material itself. Moving forward, the regenerative mode of action of naturally-derived materials requires additional investigation. Elucidating such mechanisms will allow for the determination of critical design parameters to further enhance efficacy and capitalize on the full potential of naturally-derived scaffolds.
Collapse
Affiliation(s)
- Ashley B Allen
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA, 30332, USA,
| | | | | | | |
Collapse
|
17
|
Abstract
Chronic renal failure is an important clinical problem with significant socioeconomic impact worldwide. Despite advances in renal replacement therapies and organ transplantation, poor quality of life for dialysis patients and long transplant waiting lists remain major concerns for nephrologists treating this condition. There is therefore a pressing need for novel therapies to promote renal cellular repair and tissue remodeling. Over the past decade, advances in the field of regenerative medicine allowed development of cell therapies suitable for kidney repair. Mesenchymal stem cells (MSCs) are undifferentiated cells that possess immunomodulatory and tissue trophic properties and the ability to differentiate into multiple cell types. Studies in animal models of chronic renal failure have uncovered a unique potential of these cells for improving function and regenerating the damaged kidney. Nevertheless, several limitations pertaining to inadequate engraftment, difficulty to monitor, and untoward effects of MSCs remain to be addressed. Adverse effects observed following intravascular administration of MSCs include immune rejection, adipogenic differentiation, malignant transformation, and prothrombotic events. Nonetheless, most studies indicate a remarkable capability of MSCs to achieve kidney repair. This review summarizes the regenerative potential of MSCs to provide functional recovery from renal failure, focusing on their application and the current challenges facing clinical translation.
Collapse
|
18
|
Cowan PJ, Cooper DKC, d'Apice AJF. Kidney xenotransplantation. Kidney Int 2014; 85:265-75. [PMID: 24088952 PMCID: PMC3946635 DOI: 10.1038/ki.2013.381] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/12/2013] [Accepted: 07/17/2013] [Indexed: 12/14/2022]
Abstract
Xenotransplantation using pigs as donors offers the possibility of eliminating the chronic shortage of donor kidneys, but there are several obstacles to be overcome before this goal can be achieved. Preclinical studies have shown that, while porcine renal xenografts are broadly compatible physiologically, they provoke a complex rejection process involving preformed and elicited antibodies, heightened innate immune cell reactivity, dysregulated coagulation, and a strong T cell-mediated adaptive response. Furthermore, the susceptibility of the xenograft to proinflammatory and procoagulant stimuli is probably increased by cross-species molecular defects in regulatory pathways. To balance these disadvantages, xenotransplantation has at its disposal a unique tool to address particular rejection mechanisms and incompatibilities: genetic modification of the donor. This review focuses on the pathophysiology of porcine renal xenograft rejection, and on the significant genetic, pharmacological, and technical progress that has been made to prolong xenograft survival.
Collapse
Affiliation(s)
- Peter J Cowan
- 1] Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia [2] Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anthony J F d'Apice
- 1] Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia [2] Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Abstract
The shortage of human organs for transplantation has focused research on the possibility of transplanting pig organs into humans. Many factors contribute to the failure of a pig organ graft in a primate. A rapid innate immune response (natural anti-pig antibody, complement activation, and an innate cellular response; e.g., neutrophils, monocytes, macrophages, and natural killer cells) is followed by an adaptive immune response, although T-cell infiltration of the graft has rarely been reported. Other factors (e.g., coagulation dysregulation and inflammation) appear to play a significantly greater role than in allotransplantation. The immune responses to a pig xenograft cannot therefore be controlled simply by suppression of T-cell activity. Before xenotransplantation can be introduced successfully into the clinic, the problems of the innate, coagulopathic, and inflammatory responses will have to be overcome, most likely by the transplantation of organs from genetically engineered pigs. Many of the genetic manipulations aimed at protecting against these responses also reduce the adaptive response. The T-cell and elicited antibody responses can be prevented by the biological and/or pharmacologic agents currently available, in particular, by costimulation blockade-based regimens. The exogenous immunosuppressive regimen may be significantly reduced by the presence of a graft from a pig transgenic for a mutant (human) class II transactivator gene, resulting in down-regulation of swine leukocyte antigen class II expression, or from a pig with "local" vascular endothelial cell expression of an immunosuppressive gene (e.g., CTLA4-Ig). The immunomodulatory efficacy of regulatory T cells or mesenchymal stromal cells has been demonstrated in vitro but not yet in vivo.
Collapse
|
20
|
Brückner S, Tautenhahn HM, Winkler S, Stock P, Dollinger M, Christ B. A fat option for the pig: hepatocytic differentiated mesenchymal stem cells for translational research. Exp Cell Res 2013; 321:267-75. [PMID: 24200501 DOI: 10.1016/j.yexcr.2013.10.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/21/2013] [Accepted: 10/27/2013] [Indexed: 02/07/2023]
Abstract
STUDY BACKGROUND Extended liver resection is the only curative treatment option of liver cancer. Yet, the residual liver may not accomplish the high metabolic and regenerative capacity needed, which frequently leads to acute liver failure. Because of their anti-inflammatory and -apoptotic as well as pro-proliferative features, mesenchymal stem cells differentiated into hepatocyte-like cells might provide functional and regenerative compensation. Clinical translation of basic research requires pre-clinical approval in large animals. Therefore, we characterized porcine mesenchymal stem cells (MSC) from adipose tissue and bone marrow and their hepatocyte differentiation potential for future assessment of functional liver support after surgical intervention in the pig model. METHODS Mesenchymal surface antigens and multi-lineage differentiation potential of porcine MSC isolated by collagenase digestion either from bone marrow or adipose tissue (subcutaneous/visceral) were assessed by flow cytometry. Morphology and functional properties (urea-, glycogen synthesis and cytochrome P450 activity) were determined during culture under differentiation conditions and compared with primary porcine hepatocytes. RESULTS MSC from porcine adipose tissue and from bone marrow express the typical mesenchymal markers CD44, CD29, CD90 and CD105 but not haematopoietic markers. MSC from both sources displayed differentiation into the osteogenic as well as adipogenic lineage. After hepatocyte differentiation, expression of CD105 decreased significantly and cells adopted the typical polygonal morphology of hepatocytes. Glycogen storage was comparable in adipose tissue- and bone marrow-derived cells. Urea synthesis was about 35% lower in visceral than in subcutaneous adipose tissue-derived MSC. Cytochrome P450 activity increased significantly during differentiation and was twice as high in hepatocyte-like cells generated from bone marrow as from adipose tissue. CONCLUSION The hepatocyte differentiation of porcine adipose tissue-derived MSC was shown for the first time yielding hepatocyte-like cells with specific functions similar in bone marrow and subcutaneous adipose tissue-derived MSC. That makes them good pre-clinical candidates for supportive approaches after liver resection in the pig.
Collapse
Affiliation(s)
- Sandra Brückner
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Hans-Michael Tautenhahn
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany; TRM, Translational Centre for Regenerative Medicine, Philipp-Rosenthal-Str. 55, Leipzig D-04103, Germany.
| | - Sandra Winkler
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Peggy Stock
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Matthias Dollinger
- University Hospital Ulm, First Department of Medicine, Albert-Einstein-Allee 23, Ulm D-89081, Germany.
| | - Bruno Christ
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany; TRM, Translational Centre for Regenerative Medicine, Philipp-Rosenthal-Str. 55, Leipzig D-04103, Germany.
| |
Collapse
|
21
|
Li J, Andreyev O, Chen M, Marco M, Iwase H, Long C, Ayares D, Shen Z, Cooper DKC, Ezzelarab MB. Human T cells upregulate CD69 after coculture with xenogeneic genetically-modified pig mesenchymal stromal cells. Cell Immunol 2013; 285:23-30. [PMID: 24044963 DOI: 10.1016/j.cellimm.2013.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/24/2013] [Accepted: 08/20/2013] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSC) obtained from α1,3-galactosyltransferase gene knock-out pigs transgenic for the human complement-regulatory protein CD46 (GTKO/CD46 pMSC) suppress in vitro human anti-pig cellular responses as efficiently as allogeneic human MSC. We investigated the immunoregulatory effects of GTKO/CD46 pMSC on human CD4(+) and CD8(+) T cell proliferation in response to pig aortic endothelial cells (pAEC). pMSC efficiently suppressed T cell proliferation, which was associated with downregulation of granzyme B expression. No induction of CD4(+)CD25(+)Foxp3(hi) regulatory T cells or T cell apoptosis was documented. In correlation with T cell proliferation, CD25 expression was upregulated on T cells in response to pAEC but not to pMSC. In contrast, CD69 expression was upregulated on T cells in response to both pMSC and pAEC, which was associated with a significant increase in the phosphorylation of STAT5. GTKO/CD46 pMSC possibly regulate human T cell responses through modulation of CD69 expression and STAT5 signaling.
Collapse
Affiliation(s)
- Jiang Li
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Transplantation Surgery, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Expansion of multipotent stem cells from the adult human brain. PLoS One 2013; 8:e71334. [PMID: 23967194 PMCID: PMC3743777 DOI: 10.1371/journal.pone.0071334] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 06/30/2013] [Indexed: 12/22/2022] Open
Abstract
The discovery of stem cells in the adult human brain has revealed new possible scenarios for treatment of the sick or injured brain. Both clinical use of and preclinical research on human adult neural stem cells have, however, been seriously hampered by the fact that it has been impossible to passage these cells more than a very few times and with little expansion of cell numbers. Having explored a number of alternative culturing conditions we here present an efficient method for the establishment and propagation of human brain stem cells from whatever brain tissue samples we have tried. We describe virtually unlimited expansion of an authentic stem cell phenotype. Pluripotency proteins Sox2 and Oct4 are expressed without artificial induction. For the first time multipotency of adult human brain-derived stem cells is demonstrated beyond tissue boundaries. We characterize these cells in detail in vitro including microarray and proteomic approaches. Whilst clarification of these cells' behavior is ongoing, results so far portend well for the future repair of tissues by transplantation of an adult patient's own-derived stem cells.
Collapse
|
23
|
Li J, Ezzelarab MB, Cooper DKC. Do mesenchymal stem cells function across species barriers? Relevance for xenotransplantation. Xenotransplantation 2013; 19:273-85. [PMID: 22978461 DOI: 10.1111/xen.12000] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Allogeneic mesenchymal stem (stromal) cells (MSC) are a promising therapy for various pathological conditions. Genetically modified pig MSC have been demonstrated to downregulate the human T-cell response to pig antigens in vitro. Before genetically modified pig MSC can be used clinically, however, evidence needs to be provided to indicate whether they will survive in a human (xenogeneic) host. LITERATURE SEARCH AND RESULTS A literature search through the end of 2011 identified 94 reports of the in vivo cross-species administration of MSC in a variety of experimental models. The majority (n = 89) involved the use of human MSC in various other species, with an occasional study using pig, rat, or guinea-pig MSC. When human MSC were used, they were largely derived from the bone marrow, adipose tissue, or umbilical cord blood. The routes of administration were varied, although almost half of the studies utilized the intravenous route. In 88 experiments (93.6%), there was evidence that the MSC engrafted and functioned across the species barrier, and in only six cases (6.4%) was there evidence of failure to function. Importantly, MSC function was confirmed in several different cross-species models. For example, human MSC functioned in no fewer than seven different recipient species. CONCLUSIONS The data provided by this literature search strengthen the hypothesis that pig MSC will function satisfactorily in a different species, for example, humans. The data also suggest that our own in vitro observations on the efficacy of pig MSC in downregulating the strength of the human T-cell response to pig antigens will likely be reproduced in vivo in pre-clinical large animal models and in clinical trials.
Collapse
Affiliation(s)
- Jiang Li
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
24
|
Schneider MKJ, Seebach JD. Xenotransplantation literature update, March to April 2012. Xenotransplantation 2012; 19:207-11. [PMID: 22702472 DOI: 10.1111/j.1399-3089.2012.00707.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mårten K J Schneider
- Division of Internal Medicine, University Hospital Zurich Service of Immunology and Allergology, Department of Internal Medicine, University Hospital and Medical Faculty, Geneva, Switzerland.
| | | |
Collapse
|