1
|
Reid G, Cerino G, Melly L, Fusco D, Zhang C, Reuthebuch O, Milan G, Marsano A. Harnessing the angiogenic potential of adipose-derived stromal vascular fraction cells with perfusion cell seeding. Stem Cell Res Ther 2025; 16:220. [PMID: 40312732 PMCID: PMC12044990 DOI: 10.1186/s13287-025-04286-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The rapid formation and long-term maintenance of functional vascular networks are crucial for the success of regenerative therapies. The stromal vascular fraction (SVF) from human adipose tissue is a readily available, heterogeneous cell source containing myeloid lineage cells, mesenchymal stromal cells, endothelial cells and their precursors, and pericytes, which are important for vascular support. Previous studies showed that seeding SVF cells under perfusion and pre-culturing them on three-dimensional (3D) collagen sponges enhances the vascular cell component in vitro while accelerating vascularization and improving human cell engraftment in vivo compared to static pre-culture. However, generating a perfusion-cultured SVF patch over a 5-day period is both costly and challenging for clinical translation. To overcome these limitations, this study explores a no-pre-culture strategy by comparing perfusion-based seeding with static cell loading on 3D sponges. The hypothesis is that perfusion-based seeding enhances in vivo cell engraftment and angiogenic potential by loading different SVF cell subpopulations onto 3D scaffolds during the seeding process. METHODS SVF-cells are seeded onto collagen scaffold using two approaches: a closed system perfusion bioreactor for 18 h or static loading onto the sponge surface. The in vitro cell distribution and baseline cytokine profiles were evaluated. Subsequently, human cell engraftment and differentiation were assessed in vivo using a nude rat subcutaneous implantation model. Analyses included the survival of transplanted human cells, the functionality and maturation of newly formed blood vessels within the SVF-patch. RESULTS Perfusion seeding significantly reduced the number of myeloid cells and achieved uniform spatial distribution across the construct. Vascular endothelial growth factor release was significantly increased following perfusion culture, whereas pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-1β were decreased. In the short term, perfusion culture enhanced uniform vascularization and SVF cell engraftment in vivo. However, the long-term differences between the perfusion-seeded and static-seeded groups diminished. CONCLUSION Eliminating the need for prolonged pre-culture offers a feasible and cost-effective strategy for advancing regenerative cell-based therapies by reducing pre-culture times while preserving therapeutic efficacy. Perfusion-based seeding offers significant short-term benefits, including enhanced vascularization and cell engraftment, though long-term differences compared to static seeding are minimal. Further investigation is needed to evaluate its potential in a diseased ischemic heart model.
Collapse
Affiliation(s)
- Gregory Reid
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
- Department of Plastic and Hand Surgery, University Hospital of Zürich, 8091, Zurich, Switzerland
| | - Giulia Cerino
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Ludovic Melly
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Deborah Fusco
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Chunyan Zhang
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Oliver Reuthebuch
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Giulia Milan
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Anna Marsano
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland.
| |
Collapse
|
2
|
Lee HY, An SB, Hwang SY, Hwang GY, Lee HL, Park HJ, Shin J, Kim KN, Wee SW, Yoon SL, Ha Y. Synergistic enhancement of spinal fusion in preclinical models using low-dose rhBMP-2 and stromal vascular fraction in an injectable hydrogel composite. Mater Today Bio 2025; 30:101379. [PMID: 39759847 PMCID: PMC11699625 DOI: 10.1016/j.mtbio.2024.101379] [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: 09/11/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Spinal fusion surgery remains a significant challenge due to limitations in current bone graft materials, particularly in terms of bioactivity, integration, and safety. This study presents an innovative approach using an injectable hydroxyapatite/β-tricalcium phosphate (HA/β-TCP) hydrogel combined with stromal vascular fraction (SVF) and low-dose recombinant human BMP-2 (rhBMP-2) to enhance osteodifferentiation and angiogenesis. Through a series of in vitro studies and preclinical models involving rats and minipigs, we demonstrated that the hydrogel system enables the sustained release of rhBMP-2, resulting in significantly improved bone density and integration, alongside reduced inflammatory responses. The combination of rhBMP-2 and SVF in this injectable formulation yielded superior spinal fusion outcomes, with enhanced mechanical properties and increased bone mass in both small and large animal models. These findings suggest that this strategy offers a promising and safer alternative for spinal fusion, with strong potential for clinical application.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Graduate School, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Jung Park
- Department of Research Center, CGBio., co. Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Joongkyum Shin
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung Won Wee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sol Lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| |
Collapse
|
3
|
Adipose Tissue and Adipose-Tissue-Derived Cell Therapies for the Treatment of the Face and Hands of Patients Suffering from Systemic Sclerosis. Biomedicines 2023; 11:biomedicines11020348. [PMID: 36830886 PMCID: PMC9953720 DOI: 10.3390/biomedicines11020348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/28/2023] Open
Abstract
Adipose tissue is recognized as a valuable source of cells with angiogenic, immunomodulatory, reparative and antifibrotic properties and emerged as a therapeutic alternative for the regeneration and repair of damaged tissues. The use of adipose-tissue-based therapy is expanding in autoimmune diseases, particularly in Systemic Sclerosis (SSc), a disease in which hands and face are severely affected, leading to disability and a decrease in quality of life. Combining the advantage of an abundant supply of fat tissue and a high abundance of stem/stromal cells, fat grafting and adipose tissue-derived cell-based therapies are attractive therapeutic options in SSc. This review aims to synthesize the evidence to determine the effects of the use of these biological products for face and hands treatment in the context of SSc. This highlights several points: the need to use relevant effectiveness criteria taking into account the clinical heterogeneity of SSc in order to facilitate assessment and comparison of innovative therapies; second, it reveals some impacts of the disease on fat-grafting success; third, an important heterogeneity was noticed regarding the manufacturing of the adipose-derived products and lastly, it shows a lack of robust evidence from controlled trials comparing adipose-derived products with standard care.
Collapse
|
4
|
Intraoperative Administration of Adipose Stromal Vascular Fraction Does Not Improve Functional Outcomes in Young Patients with Anterior Cruciate Ligament Reconstruction. J Clin Med 2022; 11:jcm11216240. [PMID: 36362468 PMCID: PMC9654782 DOI: 10.3390/jcm11216240] [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: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Adipose stromal vascular fraction (SVF) has a versatile cellular system for biologically augmented therapies. However, there have been no clinical studies investigating the benefits of the augmentation of anterior cruciate ligament reconstruction (ACLR) with SVF. We conducted a retrospective study in assessing the effects of intraoperative SVF administration on the functional outcomes in young patients with ACLR. The enrolled patients were divided into the control group (ACLR only) and the SVF group (ACLR with SVF). The functional outcomes in both groups were assessed by the Lysholm knee scoring system, the Tegner activity scale, and the International Knee Documentation Committee (IKDC) subjective evaluation form, and compared at several time points during a 12-month follow-up. We found that the sex distribution and pre-surgery scores were similar in the two groups, whereas the mean age of the SVF group was higher than that of the control group (p = 0.046). The between-group analysis and generalized estimating equation model analysis revealed that, while patients in the SVF group significantly improved all their functional outcomes at 12 months after surgery, this improvement was not significantly different from the results of patients in the control group (Lysholm, p = 0.553; Tegner, p = 0.197; IKDC, p = 0.486). No side effects were observed in either group. We concluded that the intraoperative administration of SVF does not improve or accelerate functional recovery after ACLR in young patients.
Collapse
|
5
|
Hodges NA, Lampejo AO, Shang H, Rowe G, LeBlanc AJ, Katz AJ, Murfee WL. Viewing stromal vascular fraction de novo vessel formation and association with host microvasculature using the rat mesentery culture model. Microcirculation 2022; 29:e12758. [PMID: 35466504 PMCID: PMC9592675 DOI: 10.1111/micc.12758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 03/26/2022] [Accepted: 04/22/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The objective of the study is to demonstrate the innovation and utility of mesenteric tissue culture for discovering the microvascular growth dynamics associated with adipose-derived stromal vascular fraction (SVF) transplantation. Understanding how SVF cells contribute to de novo vessel growth (i.e., neovascularization) and host network angiogenesis motivates the need to make observations at single-cell and network levels within a tissue. METHODS Stromal vascular fraction was isolated from the inguinal adipose of adult male Wistar rats, labeled with DiI, and seeded onto adult Wistar rat mesentery tissues. Tissues were then cultured in MEM + 10% FBS for 3 days and labeled for BSI-lectin to identify vessels. Alternatively, SVF and tissues from green fluorescent-positive (GFP) Sprague Dawley rats were used to track SVF derived versus host vasculature. RESULTS Stromal vascular fraction-treated tissues displayed a dramatically increased vascularized area compared to untreated tissues. DiI and GFP+ tracking of SVF identified neovascularization involving initial segment formation, radial outgrowth from central hub-like structures, and connection of segments. Neovascularization was also supported by the formation of segments in previously avascular areas. New segments characteristic of SVF neovessels contained endothelial cells and pericytes. Additionally, a subset of SVF cells displayed the ability to associate with host vessels and the presence of SVF increased host network angiogenesis. CONCLUSIONS The results showcase the use of the rat mesentery culture model as a novel tool for elucidating SVF cell transplant dynamics and highlight the impact of model selection for visualization.
Collapse
Affiliation(s)
- Nicholas A. Hodges
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Arinola O. Lampejo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Hulan Shang
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Gabrielle Rowe
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | - Amanda Jo LeBlanc
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | - Adam J. Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Walter L. Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
Rowe G, Heng DS, Beare JE, Hodges NA, Tracy EP, Murfee WL, LeBlanc AJ. Stromal Vascular Fraction Reverses the Age-Related Impairment in Revascularization following Injury. J Vasc Res 2022; 59:343-357. [PMID: 36075199 PMCID: PMC9780192 DOI: 10.1159/000526002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/06/2022] [Indexed: 12/31/2022] Open
Abstract
Adipose-derived stromal vascular fraction (SVF) has emerged as a potential regenerative therapy, but few studies utilize SVF in a setting of advanced age. Additionally, the specific cell population in SVF providing therapeutic benefit is unknown. We hypothesized that aging would alter the composition of cell populations present in SVF and its ability to promote angiogenesis following injury, a mechanism that is T cell-mediated. SVF isolated from young and old Fischer 344 rats was examined with flow cytometry for cell composition. Mesenteric windows from old rats were isolated following exteriorization-induced (EI) hypoxic injury and intravenous injection of one of four cell therapies: (1) SVF from young or (2) old donors, (3) SVF from old donors depleted of or (4) enriched for T cells. Advancing age increased the SVF T-cell population but reduced revascularization following injury. Both young and aged SVF incorporated throughout the host mesenteric microvessels, but only young SVF significantly increased vascular area following EI. This study highlights the effect of donor age on SVF angiogenic efficacy and demonstrates how the ex vivo mesenteric-window model can be used in conjunction with SVF therapy to investigate its contribution to angiogenesis.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA,
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA,
| | - David S Heng
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Nicholas A Hodges
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Evan P Tracy
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Baboudjian M, Gondran-Tellier B, Boissier R, Ancel P, Marjollet J, Lyonnet L, François P, Sabatier F, Lechevallier E, Dutour A, Paul P. An enhanced level of VCAM in transplant preservation fluid is an independent predictor of early kidney allograft dysfunction. Front Immunol 2022; 13:966951. [PMID: 36032101 PMCID: PMC9403542 DOI: 10.3389/fimmu.2022.966951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Background We aimed to evaluate whether donor-related inflammatory markers found in kidney transplant preservation fluid can associate with early development of kidney allograft dysfunction. Methods Our prospective study enrolled 74 consecutive donated organs who underwent kidney transplantation in our center between September 2020 and June 2021. Kidneys from 27 standard criteria donors were allocated to static cold storage and kidneys from 47 extended criteria donors to hypothermic machine perfusion. ELISA assessment of inflammatory biomarkers (IL-6, IL6-R, ICAM, VCAM, TNFα, IFN-g, CXCL1 and Fractalkine) was analyzed in view of a primary endpoint defined as the occurrence of delayed graft function or slow graft function during the first week following transplantation. Results Soluble VCAM levels measured in transplant conservation fluid were significantly associated with recipient serum creatinine on day 7. Multivariate stepwise logistic regression analysis identified VCAM as an independent non-invasive predictor of early graft dysfunction, both at 1 week (OR: 3.57, p = .04, 95% CI: 1.06-12.03) and 3 Months (OR: 4.039, p = .034, 95% CI: 1.11-14.73) after transplant surgery. Conclusions This prospective pilot study suggests that pre-transplant evaluation of VCAM levels could constitute a valuable indicator of transplant health and identify the VCAM-CD49d pathway as a target to limit donor-related vascular injury of marginal transplants.
Collapse
Affiliation(s)
- Michael Baboudjian
- Department of Urology and Transplantation, La Conception Hospital, Assistance Publique-Hôpitaux Marseille, Marseille, France
- Department of Urology, Assistance Publique-Hôpitaux de Marseille, Hopital Nord, Aix-Marseille University, Marseille, France
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Bastien Gondran-Tellier
- Department of Urology and Transplantation, La Conception Hospital, Assistance Publique-Hôpitaux Marseille, Marseille, France
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Romain Boissier
- Department of Urology and Transplantation, La Conception Hospital, Assistance Publique-Hôpitaux Marseille, Marseille, France
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Patricia Ancel
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Juline Marjollet
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Luc Lyonnet
- Department of Hematology, Hopital de la Conception, Assistance Publique-Hôpitaux Marseille, Marseille, France
| | - Pauline François
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
| | - Florence Sabatier
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
- Cell Therapy Laboratory, Centre d'Investigation Clinique (CIC)-149, La Conception Hospital, Assistance Publique-Hôpitaux Marseille, Marseille, France
| | - Eric Lechevallier
- Department of Urology and Transplantation, La Conception Hospital, Assistance Publique-Hôpitaux Marseille, Marseille, France
| | - Anne Dutour
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
- Endocrinology, Metabolic Diseases and Nutrition Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Pascale Paul
- Institut national de la santé et de la recherche médicale (INSERM) 1263, Aix Marseille University, French national research institute for agriculture, food and the environment (INRAE), Centre de recherche en CardioVasculaire et Nutrition (C2VN), Marseille, France
- Department of Hematology, Hopital de la Conception, Assistance Publique-Hôpitaux Marseille, Marseille, France
- Institut national de la santé et de la recherche médicale (INSERM) unité mixte de recherche (UMR)_1090, Aix Marseille University, TAGC Theories and Approaches of Genomic Complexity, Parc Scientifique de Luminy Case 928, Marseille, France
- *Correspondence: Pascale Paul,
| |
Collapse
|
8
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
9
|
Majbour D, Suarez-Martinez AD, Hodges NA, Lampejo AO, Lomel BM, Rice EW, Shang H, Katz AJ, Murfee WL. An Ex Vivo Tissue Culture Method for Discovering Cell Dynamics Involved in Stromal Vascular Fraction Vasculogenesis Using the Mouse Mesentery. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:157-170. [PMID: 35099735 DOI: 10.1007/978-1-0716-2059-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Stromal vascular fraction (SVF), isolated from adipose tissue, identifies as a rich cell source comprised of endothelial cells, endothelial progenitor cells, pericytes, smooth muscle cells, fibroblasts, and immune cells. SVF represents a promising therapeutic heterogonous cell source for growing new blood microvessels due to its rich niche of cells. However, the spatiotemporal dynamics of SVF within living tissues remain largely unknown. The objective of this chapter is to describe a protocol for culturing SVF on mouse mesentery tissues in order to aid in the discovery of SVF dynamics and associated vessel growth over time. SVF was isolated from the inguinal adipose from adult mice and seeded onto mesentery tissues. Tissues were then cultured for up to 5 days and labeled with endothelial cell and pericyte markers. Representative results demonstrate the observation of SVF-derived vasculogenesis characterized by de novo vessel formation and subsequent vessel connection.
Collapse
Affiliation(s)
- Dima Majbour
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Ariana D Suarez-Martinez
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nicholas A Hodges
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Arinola O Lampejo
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Banks M Lomel
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Elijah W Rice
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hulan Shang
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Adam J Katz
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Walter L Murfee
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
Zeng A, Wang SR, He YX, Yan Y, Zhang Y. Progress in understanding of the stalk and tip cells formation involvement in angiogenesis mechanisms. Tissue Cell 2021; 73:101626. [PMID: 34479073 DOI: 10.1016/j.tice.2021.101626] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/28/2022]
Abstract
Vascular sprouting is a key process of angiogenesis and mainly related to the formation of stalk and tip cells. Many studies have found that angiogenesis has a great clinical significance in promoting the functional repair of impaired tissues and anti-angiogenesis is a key to treatment of many tumors. Therefore, how the pathways regulate angiogenesis by regulating the formation of stalk and tip cells is an urgent problem for researchers. This review mainly summarizes the research progress of pathways affecting the formation of stalk and tip cells during angiogenesis in recent years, including the main signaling pathways (such as VEGF-VEGFR-Dll4-Notch signaling pathway, ALK-Smad signaling pathway,CCN1-YAP/YAZ signaling pathway and other signaling pathways) and cellular actions (such as cellular metabolisms, intercellular tension and other actions), aiming to further give the readers an insight into the mechanism of regulating the formation of stalk and tip cells during angiogenesis and provide more targets for anti-angiogenic drugs.
Collapse
Affiliation(s)
- Ao Zeng
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Shu-Rong Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yu-Xi He
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yu Yan
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yan Zhang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| |
Collapse
|
11
|
Autologous Minimally Invasive Cell-Based Therapy for Meniscal and Anterior Cruciate Ligament Regeneration. Case Rep Orthop 2021; 2021:6614232. [PMID: 34258092 PMCID: PMC8253646 DOI: 10.1155/2021/6614232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 06/02/2021] [Accepted: 06/06/2021] [Indexed: 01/16/2023] Open
Abstract
The meniscus is a fibrocartilaginous tissue that acts as a “shock absorber,” along with performing functions such as stabilization and lubrication of the joint, proprioception, and load distribution. Sudden twisting movements during weight bearing or trauma can cause injury to the menisci, which leads to symptoms such as pain, swelling, and difficulty in performing movements, among others. Conventional pharmacological and surgical treatments are effective in treating the condition; however, do not result in regeneration of healthy tissues. In this report, we highlight the role of cell-based therapy in the management of medial and lateral meniscal and anterior cruciate ligament tears in a patient who was unwilling to undergo surgical treatment. We injected autologous mesenchymal stem cells obtained from the bone marrow and adipose tissue and platelet-rich plasma into the joint of the patient at the area of injury, as well as intravenously. The results of our study corroborate with those previously reported in the literature regarding the improvement in clinical parameters and regeneration of meniscal tissue and ligament. Thus, based on previous literature and improvements noticed in our patient, cell-based therapy can be considered a safe and effective therapeutic modality in the treatment of meniscal tears and cruciate ligament injury.
Collapse
|
12
|
Carstens MH, Quintana FJ, Calderwood ST, Sevilla JP, Ríos AB, Rivera CM, Calero DW, Zelaya ML, Garcia N, Bertram KA, Rigdon J, Dos-Anjos S, Correa D. Treatment of chronic diabetic foot ulcers with adipose-derived stromal vascular fraction cell injections: Safety and evidence of efficacy at 1 year. Stem Cells Transl Med 2021; 10:1138-1147. [PMID: 33826245 PMCID: PMC8284780 DOI: 10.1002/sctm.20-0497] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/09/2021] [Accepted: 03/04/2021] [Indexed: 12/31/2022] Open
Abstract
Diabetes affects multiple systems in complex manners. Diabetic foot ulcers (DFUs) are a result of diabetes‐induced microarterial vessel disease and peripheral neuropathy. The presence of arteriosclerosis‐induced macroarterial disease can further complicate DFU pathophysiology. Recent studies suggest that mesenchymal stromal cell therapies can enhance tissue regeneration. This phase I study was designed to determine the safety and explore the efficacy of local injections of autologous adipose‐derived stromal vascular fraction (SVF) cells to treat nonhealing DFUs greater than 3 cm in diameter. Sixty‐three patients with type 2 diabetes with chronic DFU—all amputation candidates—were treated with 30 × 106 SVF cells injected in the ulcer bed and periphery and along the pedal arteries. Patients were seen at 6 and 12 months to evaluate ulcer closure. Doppler ultrasounds were performed in a subset of subjects to determine vascular structural parameters. No intervention‐related serious adverse events were reported. At 6 months, 51 subjects had 100% DFU closure, and 8 subjects had ≥75% closure. Three subjects had early amputations, and one subject died. At 12 months, 50 subjects had 100% DFU healing and 4 subjects had ≥85% healing. Five subjects died between the 6‐ and 12‐month follow‐up visits. No deaths were intervention related. Doppler studies in 11 subjects revealed increases in peak systolic velocity and pulsatility index in 33 of 33 arteries, consistent with enhanced distal arterial runoff. These results indicate that SVF can be safely used to treat chronic DFU, with evidence of efficacy (wound healing) and mechanisms of action that include vascular repair and/or angiogenesis.
Collapse
Affiliation(s)
- Michael H Carstens
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA.,Department of Surgery, Universidad Nacional de Nicaragua, León, Nicaragua
| | | | - Santos T Calderwood
- Department of Surgery, Universidad Nacional de Nicaragua, Matagalpa, Nicaragua
| | - Juan P Sevilla
- Department of Surgery, Universidad Nacional de Nicaragua, Matagalpa, Nicaragua
| | - Arlen B Ríos
- Department of Surgery, Universidad Nacional de Nicaragua, Matagalpa, Nicaragua
| | - Carlos M Rivera
- Department of Radiology, Universidad Nacional de Nicaragua, Matagalpa, Nicaragua
| | - Dorian W Calero
- Department of Radiology, Universidad Nacional de Nicaragua, León, Nicaragua
| | - María L Zelaya
- Department of Radiology, Universidad Nacional de Nicaragua, León, Nicaragua
| | - Nelson Garcia
- Department of Medicine, Universidad Nacional de Nicaragua, Matagalpa, Nicaragua
| | - Kenneth A Bertram
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Joseph Rigdon
- Department of Biostatistics and Data Science, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | | | - Diego Correa
- Diabetes Research Institute and Cellular Transplant Center, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
13
|
Jones VM, Suarez-Martinez AD, Hodges NA, Murfee WL, Llull R, Katz AJ. A clinical perspective on adipose-derived cell therapy for enhancing microvascular health and function: Implications and applications for reconstructive surgery. Microcirculation 2020; 28:e12672. [PMID: 33174272 DOI: 10.1111/micc.12672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/18/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022]
Abstract
Restoration of form and function requires apposition of tissues in the form of flaps to reconstitute local perfusion. Successful reconstruction relies on flap survival and its integration with the recipient bed. The flap's precariously perfused hypoxic areas undergo adaptive microvascular changes both internally and in connection with the recipient bed. A cell-mediated, coordinated response to hypoxia drives these adaptive processes, restoring a tissue's normoxic homeostasis via de novo vasculogenesis, sprouting angiogenesis, and stabilizing arterialization. As cells exert prolonged and coordinated effects on site, their use as biological agents merit translational consideration of sourcing angio-competent cells and delivering them to territories enduring microcirculatory acclimatization. Angio-competent cells abound in adipose tissue: a reliable, accessible, and expendable source of adipose-derived cells (ADC). When subject to enzymatic digestion and centrifugation, adipose tissue separates its various ADC: A subset of buoyant oil-dense adipocytes (the tissue's parenchymal component) accumulates on a supra-natant layer, whereas the mesenchymal component remains in the infra-natant sediment, containing the tissue's stromal vascular fraction (SVF), where angio-component cells abound. The SVF can be further manipulated, selected, or culture expanded into more specific stromal subsets (herein defined as adipose stromal cells, ASC). While promising clinical applications for ADC await clinical proof and regulatory authorization, basic science investigation is needed to elucidate the specific ADC mechanisms that influence microvascular growth, remodeling, and function following flap surgery. The objective of this article is to share the clinical perspectives of reconstructive plastic surgeons regarding the use of ADC-based therapies to help with flap tissue integration, revascularization, and wound healing. Specifically, the focus will be on considering the potential for ADC as therapeutic agents and how their clinical application motivates basic science opportunities.
Collapse
Affiliation(s)
- V Morgan Jones
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ariana D Suarez-Martinez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nicholas A Hodges
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Ramon Llull
- Department of Plastic Surgery, Hospital Quiron Salud PalmaPlanas, Palma, Spain
| | - Adam J Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
14
|
Tracy EP, Gettler BC, Zakhari JS, Schwartz RJ, Williams SK, Birla RK. 3D Bioprinting the Cardiac Purkinje System Using Human Adipogenic Mesenchymal Stem Cell Derived Purkinje Cells. Cardiovasc Eng Technol 2020; 11:587-604. [PMID: 32710379 DOI: 10.1007/s13239-020-00478-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 07/09/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE The objective of this study was to reprogram human adipogenic mesenchymal stem cells (hADMSCs) to form Purkinje cells and to use the reprogrammed Purkinje cells to bioprint Purkinje networks. METHODS hADMSCs were reprogrammed to form Purkinje cells using a multi-step process using transcription factors ETS2 and MESP1 to first form cardiac progenitor stem cells followed by SHOX2 and TBX3 to form Purkinje cells. A novel bioprinting method was developed based on Pluronic acid as the sacrificial material and type I collagen as the structural material. The reprogrammed Purkinje cells were used in conjunction with the novel bioprinting method to bioprint Purkinje networks. Printed constructs were evaluated for retention of functional protein connexin 40 (Cx40) and ability to undergo membrane potential changes in response to physiologic stimulus. RESULTS hADMSCs were successfully reprogrammed to form Purkinje cells based on the expression pattern of IRX3, IRX5, SEMA and SCN10. Reprogrammed purkinje cells were incorporated into a collagen type-1 bioink and the left ventricular Purkinje network was printed using anatomical images of the bovine Purkinje system as reference. Optimization studies demonstrated that 1.8 mg/mL type-I collagen at a seeding density of 300,000 cells per 200 µL resulted in the most functional bioprinted Purkinje networks. Furthermore, bioprinted Purkinje networks formed continuous syncytium, retained expression of vital functional gap junction protein Cx40 post-print, and exhibited membrane potential changes in response to electric stimulation and acetylcholine evaluated by DiBAC4(5), an electrically responsive dye. CONCLUSION Based on the results of this study, hADMSCs were successfully reprogrammed to form Purkinje cells and bioprinted to form Purkinje networks.
Collapse
Affiliation(s)
- Evan P Tracy
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, 302 E. Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Brian C Gettler
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, 302 E. Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Joseph S Zakhari
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, 302 E. Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Robert J Schwartz
- Stem Cell Engineering, Texas Heart Institute, 6770 Bertner Avenue, Houston, TX, 77225-0345, USA.,Department of Biology and Biochemistry, Science and Engineering Research Center, (SERC-Building 445), 3605 Cullen Blvd, Room 5004, Houston, TX, 77204-5060, USA
| | - Stuart K Williams
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, 302 E. Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Ravi K Birla
- Department of Biomedical Engineering, Science and Engineering Research Center, (SERC-Building 445), 3605 Cullen Blvd, Room 2005, Houston, TX, 77204, USA.
| |
Collapse
|
15
|
Boissier R, François P, Gondran Tellier B, Meunier M, Lyonnet L, Simoncini S, Magalon J, Legris T, Arnaud L, Giraudo L, Dignat George F, Karsenty G, Burtey S, Lechevallier E, Sabatier F, Paul P. Perirenal Adipose Tissue Displays an Age-Dependent Inflammatory Signature Associated With Early Graft Dysfunction of Marginal Kidney Transplants. Front Immunol 2020; 11:445. [PMID: 32256495 PMCID: PMC7089962 DOI: 10.3389/fimmu.2020.00445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Better understanding of the contribution of donor aging and comorbidity factors of expanded criteria donors (ECD) to the clinical outcome of a transplant is a challenge in kidney transplantation. We investigated whether the features of donor-derived stromal vascular fraction of perirenal adipose tissue (PRAT-SVF) could be indicative of the deleterious impact of the ECD microenvironment on a renal transplant. Methods: A comparative analysis of cellular components, transcriptomic and vasculogenic profiles was performed in PRAT-SVF obtained from 22 optimal donors and 31 ECD deceased donors. We then investigated whether these parameters could be associated with donor aging and early allograft dysfunction. Results: When compared with the PRAT-SVF of non-ECD donors, ECD PRAT-SVF displayed a lower proportion of stromal cells, a higher proportion of inflammatory NK cells. The global RNA sequencing approach indicated a differential molecular signature in the PRAT-SVF of ECD donors characterized by the over-expression of CXCL1 and IL1-β inflammatory transcripts. The vasculogenic activity of PRAT-SVF was highly variable but was not significantly affected in marginal donors. Periorgan recruitment of monocytes/macrophages and NK cells in PRAT-SVF was associated with donor aging. The presence of NK cell infiltrates was associated with lower PRAT-SVF angiogenic activity and with early allograft dysfunction evaluated on day 7 and at 1 month post-transplant. Conclusions: Our results indicate that human NK cell subsets are differentially recruited in the periorgan environment of aging kidney transplants. We provide novel evidence that PRAT-SVF represents a non-invasive and timely source of donor material with potential value to assess inflammatory features that impact organ quality and function.
Collapse
Affiliation(s)
- Romain Boissier
- Department of Urology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France.,C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France
| | - Pauline François
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Cell Therapy Department, La Conception University Hospital APHM, Aix-Marseille Univ., INSERM CIC 1409, Marseille, France
| | - Bastien Gondran Tellier
- Department of Urology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France.,C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France
| | - Maité Meunier
- Department of Nephrology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France
| | - Luc Lyonnet
- Department of Hematology and Vascular biology, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix Marseille Univ., Marseille, France
| | | | - Jeremy Magalon
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Cell Therapy Department, La Conception University Hospital APHM, Aix-Marseille Univ., INSERM CIC 1409, Marseille, France
| | - Tristan Legris
- Department of Nephrology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France
| | - Laurent Arnaud
- Department of Hematology and Vascular biology, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix Marseille Univ., Marseille, France
| | - Laurent Giraudo
- Cell Therapy Department, La Conception University Hospital APHM, Aix-Marseille Univ., INSERM CIC 1409, Marseille, France
| | - Françoise Dignat George
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Department of Hematology and Vascular biology, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix Marseille Univ., Marseille, France
| | - Gilles Karsenty
- Department of Urology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France
| | - Stéphane Burtey
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Department of Nephrology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France
| | - Eric Lechevallier
- Department of Urology and Renal Transplantation, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix-Marseille Univ., Marseille, France
| | - Florence Sabatier
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Cell Therapy Department, La Conception University Hospital APHM, Aix-Marseille Univ., INSERM CIC 1409, Marseille, France
| | - Pascale Paul
- C2VN, INSERM 1263, Aix-Marseille Univ, INRAE, Marseille, France.,Department of Hematology and Vascular biology, La Conception University Hospital, Assistance Publique-Hôpitaux Marseille (APHM), Aix Marseille Univ., Marseille, France
| |
Collapse
|
16
|
Nilforoushzadeh MA, Sisakht MM, Amirkhani MA, Seifalian AM, Banafshe HR, Verdi J, Nouradini M. Engineered skin graft with stromal vascular fraction cells encapsulated in fibrin–collagen hydrogel: A clinical study for diabetic wound healing. J Tissue Eng Regen Med 2020; 14:424-440. [DOI: 10.1002/term.3003] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/18/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022]
Affiliation(s)
| | - Mahsa Mollapour Sisakht
- Skin and Stem Cell Research CenterTehran University of Medical Sciences Tehran Iran
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
| | - Mohammad Amir Amirkhani
- Stem Cell and Regenerative Medicine Center of ExcellenceTehran University of Medical Sciences Tehran Iran
| | - Alexander M. Seifalian
- Nanotechnology and Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd)The London BioScience Innovation Centre London UK
| | - Hamid Reza Banafshe
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
- Physiology Research CenterKashan University of Medical Sciences Kashan Iran
| | - Javad Verdi
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Tehran Iran
| | - Mehdi Nouradini
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
| |
Collapse
|
17
|
Bone Tissue Regeneration in the Oral and Maxillofacial Region: A Review on the Application of Stem Cells and New Strategies to Improve Vascularization. Stem Cells Int 2019; 2019:6279721. [PMID: 32082383 PMCID: PMC7012224 DOI: 10.1155/2019/6279721] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Bone tissue engineering techniques are a promising alternative for the use of autologous bone grafts to reconstruct bone defects in the oral and maxillofacial region. However, for successful bone regeneration, adequate vascularization is a prerequisite. This review presents and discusses the application of stem cells and new strategies to improve vascularization, which may lead to feasible clinical applications. Multiple sources of stem cells have been investigated for bone tissue engineering. The stromal vascular fraction (SVF) of human adipose tissue is considered a promising single source for a heterogeneous population of essential cells with, amongst others, osteogenic and angiogenic potential. Enhanced vascularization of tissue-engineered grafts can be achieved by different mechanisms: vascular ingrowth directed from the surrounding host tissue to the implanted graft, vice versa, or concomitantly. Vascular ingrowth into the implanted graft can be enhanced by (i) optimizing the material properties of scaffolds and (ii) their bioactivation by incorporation of growth factors or cell seeding. Vascular ingrowth directed from the implanted graft towards the host tissue can be achieved by incorporating the graft with either (i) preformed microvascular networks or (ii) microvascular fragments (MF). The latter may have stimulating actions on both vascular ingrowth and outgrowth, since they contain angiogenic stem cells like SVF, as well as vascularized matrix fragments. Both adipose tissue-derived SVF and MF are cell sources with clinical feasibility due to their large quantities that can be harvested and applied in a one-step surgical procedure. During the past years, important advancements of stem cell application and vascularization in bone tissue regeneration have been made. The development of engineered in vitro 3D models mimicking the bone defect environment would facilitate new strategies in bone tissue engineering. Successful clinical application requires innovative future investigations enhancing vascularization.
Collapse
|
18
|
Moon KC, Chung HY, Han SK, Jeong SH, Dhong ES. Tissue-engineered dermis grafts using stromal vascular fraction cells on the nose: A retrospective case-control study. J Plast Reconstr Aesthet Surg 2019; 73:965-974. [PMID: 31902623 DOI: 10.1016/j.bjps.2019.11.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/29/2019] [Accepted: 11/22/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND In a previous study, our group demonstrated that cultured autologous fibroblast-seeded artificial dermis was superior to artificial dermis for covering defects after surgical excision of basal cell carcinoma (BCC) in terms of scar quality. However, utilizing cultured cells for clinical purposes requires Food and Drug Administration-approved facilities and techniques and a lengthy culture period. The purpose of this retrospective study was to compare the effects of tissue-engineered dermis containing stromal vascular fraction (SVF) cells with artificial dermis on scar quality after surgical excision of BCC on the nose. METHODS Between April 2010 and February 2018, patients who were treated with tissue-engineered or artificial dermis grafts and those with a follow-up period of greater than a year were included in this study. The Patient and Observer Scar Assessment Scales (POSAS) were compared between two groups according to the location of the graft, which was classified based on nasal subunits: the upper two-thirds zone; the lower one-third zone, except for the ala; and the alar zone. RESULTS A tissue-engineered dermis composed of SVF cells and an artificial dermis were applied to 30 and 47 patients, respectively. In upper two-thirds and lower one-third zones, except for the ala, no statistically significant differences were found in any parameters. In the alar zone, statistically significant differences were detected in 10 of 21 POSAS parameters. CONCLUSION To cover nasal defects, the tissue-engineered dermis graft may be superior to the artificial dermis graft regarding scar quality at the ala. However, there were no significant differences in other zones.
Collapse
Affiliation(s)
- Kyung-Chul Moon
- Department of Plastic Surgery, Korea University College of Medicine, 148 Guro-Dong, Guro-Ku, 152-703 Seoul, Republic of Korea
| | - Ha-Yoon Chung
- Department of Plastic Surgery, Korea University College of Medicine, 148 Guro-Dong, Guro-Ku, 152-703 Seoul, Republic of Korea
| | - Seung-Kyu Han
- Department of Plastic Surgery, Korea University College of Medicine, 148 Guro-Dong, Guro-Ku, 152-703 Seoul, Republic of Korea.
| | - Seong-Ho Jeong
- Department of Plastic Surgery, Korea University College of Medicine, 148 Guro-Dong, Guro-Ku, 152-703 Seoul, Republic of Korea
| | - Eun-Sang Dhong
- Department of Plastic Surgery, Korea University College of Medicine, 148 Guro-Dong, Guro-Ku, 152-703 Seoul, Republic of Korea
| |
Collapse
|
19
|
Karina, Samudra MF, Rosadi I, Afini I, Widyastuti T, Sobariah S, Remelia M, Puspitasari RL, Rosliana I, Tunggadewi TI. Combination of the stromal vascular fraction and platelet-rich plasma accelerates the wound healing process: pre-clinical study in a Sprague-Dawley rat model. Stem Cell Investig 2019; 6:18. [PMID: 31463311 DOI: 10.21037/sci.2019.06.08] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/02/2019] [Indexed: 12/23/2022]
Abstract
Background Adipose-derived stem cells (ADSCs) improve wound healing owing to their angiogenic potency. However, the production of large quantities of ADSCs for clinical applications is laborious. In this study, the efficacy of the stromal vascular fraction (SVF; non-expanded ADSCs) combined with platelet-rich plasma (PRP) which contains abundant growth factors, for wound healing was evaluated using an animal model. Methods PRP from venous blood and SVF from lipoaspirates were harvested from six donors. PRP, SVF, SVF + PRP, and saline solution as a negative control were injected to second degree burn wounds in the backs of 24 male Sprague-Dawley rats. On the seventh day after injection, rats were euthanized and wounds were analyzed microscopically and macroscopically. Results Wounds closed faster in the SVF + PRP group than in the control group or PRP or SVF alone groups, with less inflammation, prominent signs of re-epithelization, more skin appendages and blood vessels, and a higher rate of hair growth. No infection or rat death occurred during the trial. Conclusions The combination of SVF and PRP may provide an additive stimulatory effect to support angiogenesis and accelerate the wound healing process; accordingly, this combination is a potential alternative to ADSC treatment.
Collapse
Affiliation(s)
- Karina
- Doctoral Program of Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Klinik Hayandra, Yayasan Hayandra Peduli, Jakarta, Indonesia.,HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | | | - Imam Rosadi
- HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia.,Master Degree of Biotechnology, Institut Teknologi Bandung, Bandung, Indonesia
| | - Irsyah Afini
- HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | | | - Siti Sobariah
- HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | - Melinda Remelia
- Doctoral Program of Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Department of Basic Biomedicine, Faculty of Medicine, Universitas Kristen Indonesia, Jakarta, Indonesia
| | | | - Iis Rosliana
- HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | - Tri I Tunggadewi
- Animal Hospital Faculty of Veterinary Medicine, Bogor Agricultural Institute, Bogor, Indonesia
| |
Collapse
|
20
|
Rowe G, Kelm NQ, Beare JE, Tracy E, Yuan F, LeBlanc AJ. Enhanced beta-1 adrenergic receptor responsiveness in coronary arterioles following intravenous stromal vascular fraction therapy in aged rats. Aging (Albany NY) 2019; 11:4561-4578. [PMID: 31296794 PMCID: PMC6660031 DOI: 10.18632/aging.102069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/25/2019] [Indexed: 04/30/2023]
Abstract
Our past study showed that a single tail vein injection of adipose-derived stromal vascular fraction (SVF) into old rats was associated with improved dobutamine-mediated coronary flow reserve. We hypothesize that i.v. injection of SVF improves coronary microvascular function in aged rats via alterations in beta adrenergic microvascular signaling. Female Fischer-344 rats aged young (3 months, n=32) and old (24 months, n=30) were utilized, along with two cell therapies intravenously injected in old rats four weeks prior to sacrifice: 1x107 green fluorescent protein (GFP+) SVF cells (O+SVF, n=21), and 5x106 GFP+ bone-marrow mesenchymal stromal cells (O+BM, n=6), both harvested from young donors. Cardiac ultrasound and pressure-volume measurements were obtained, and coronary arterioles were isolated from each group for microvessel reactivity studies and immunofluorescence staining. Coronary flow reserve decreased with advancing age, but this effect was rescued by the SVF treatment in the O+SVF group. Echocardiography showed an age-related diastolic dysfunction that was improved with SVF to a greater extent than with BM treatment. Coronary arterioles isolated from SVF-treated rats showed amelioration of the age-related decrease in vasodilation to a non-selective β-AR agonist. I.v. injected SVF cells improved β-adrenergic receptor-dependent coronary flow and microvascular function in a model of advanced age.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| | - Natia Q. Kelm
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
| | - Jason E. Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA
| | - Evan Tracy
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| | - Fangping Yuan
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
| | - Amanda J. LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
21
|
Rigotti G, Chirumbolo S. Biological Morphogenetic Surgery: A Minimally Invasive Procedure to Address Different Biological Mechanisms. Aesthet Surg J 2019; 39:745-755. [PMID: 30137183 DOI: 10.1093/asj/sjy198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We present a methodology called biological morphogenetic surgery (BMS) that can recover (enlarge or reduce) the shape/volume of anatomic structures/tissues affected by congenital or acquired malformations based on a minimally invasive procedure. This emerges as a new concept in which the main task of surgery is the biological modulation of different remodeling and repair mechanisms. When applied, for example, to a tuberous breast deformity, the "enlarging BMS" expands the retracted tissue surrounding the gland through a cutting tip of a needle being inserted through small incisions percutaneously, accounting for the biological activity of the grafted fat. The obtained spaces might be spontaneously occupied and later filled with autologous grafted fat, which promotes tissue expansion by eliciting adipogenesis and preventing fibrosis. The "reducing BMS" creates an interruption of the contact between the derma and the hypoderma of the abnormally large areola and then promotes adipocytes to induce a fibrotic reaction, leading to areola reduction. Current evidence suggests that BMS might induce a bivalent mesenchymalization of the adipocyte, which promotes either new adipogenesis and angiogenesis of local fat (expanding BMS) or the granulation tissue/fibrotic response (reducing BMS), thus leading to the physiological recovery of the affected structures/tissues to normality. Level of Evidence: 4.
Collapse
Affiliation(s)
- Gino Rigotti
- Unit Head of Reconstructive Breast and Plastic Surgery, Clinica San Francesco, Verona, Italy
| | - Salvatore Chirumbolo
- Department of Neuroscience, Biomedicine and Movement Sciences-University of Verona, Verona, Italy
| |
Collapse
|
22
|
Shanbhag S, Suliman S, Pandis N, Stavropoulos A, Sanz M, Mustafa K. Cell therapy for orofacial bone regeneration: A systematic review and meta-analysis. J Clin Periodontol 2019; 46 Suppl 21:162-182. [DOI: 10.1111/jcpe.13049] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/17/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Siddharth Shanbhag
- Department of Clinical Dentistry; Center for Clinical Dental Research; University of Bergen; Bergen Norway
| | - Salwa Suliman
- Department of Clinical Dentistry; Center for Clinical Dental Research; University of Bergen; Bergen Norway
| | - Nikolaos Pandis
- Department of Orthodontics and Dentofacial Orthopedics; University of Bern; Bern Switzerland
| | - Andreas Stavropoulos
- Department of Periodontology; Faculty of Odontology; Malmö University; Malmö Sweden
| | - Mariano Sanz
- Section of Periodontology; Faculty of Odontology; University Complutense of Madrid; Madrid Spain
| | - Kamal Mustafa
- Department of Clinical Dentistry; Center for Clinical Dental Research; University of Bergen; Bergen Norway
| |
Collapse
|
23
|
Moon KC, Chung HY, Han SK, Jeong SH, Dhong ES. Possibility of Injecting Adipose-Derived Stromal Vascular Fraction Cells to Accelerate Microcirculation in Ischemic Diabetic Feet: A Pilot Study. Int J Stem Cells 2019; 12:107-113. [PMID: 30836733 PMCID: PMC6457712 DOI: 10.15283/ijsc18101] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 12/25/2022] Open
Abstract
Background and Objectives Beneficial effects of human adipose-derived stromal vascular fraction (SVF) cell injection on microcirculation have been recently reported in in vitro and in vivo studies. However, no clinical studies have reported its effect in diabetic patients who commonly experience compromised tissue perfusion, regardless of the status of intravascular blood flow. The present piloting study was designed to clinically examine the possibility of SVF cell injection to accelerate microcirculation, particularly in ischemic diabetic feet. Methods Ten diabetic feet were included to receive subcutaneous injection of SVF cells around wounds. Transcutaneous partial oxygen pressure (TcPO2) and cutaneous microvascular blood flow were measured before and every four weeks after cell injection until the 12th week visit. Results TcPO2 values increased from 31.3±7.4 before injection to 46.4±8.2 mmHg at 12 weeks after SVF injection (1.5-fold, p<0.05). Cutaneous microvascular blood flow levels increased from 34.0±21.1 before injection to 76.1±32.5 perfusion unit at 12 weeks after SVF injection (2.2-fold, p<0.05). There were no adverse events related to SVF cell injection. Conclusions Results of this study demonstrate that adipose-derived SVF cell injection have the possibility to provide beneficial effects on microcirculation in ischemic diabetic feet.
Collapse
Affiliation(s)
- Kyung-Chul Moon
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Ha-Yoon Chung
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Seung-Kyu Han
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Seong-Ho Jeong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Eun-Sang Dhong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|