1
|
Trotzier C, Bellanger C, Abdessadeq H, Delannoy P, Mojallal A, Auxenfans C. Deciphering influence of donor age on adipose-derived stem cells: in vitro paracrine function and angiogenic potential. Sci Rep 2024; 14:27589. [PMID: 39528480 PMCID: PMC11555058 DOI: 10.1038/s41598-024-73875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND As fat grafting is commonly used as a filler, Adipose-derived stem/stromal cells (ASC) have been reported to be key player in retention rate. Paracrine and differentiation potential of those cells confer them strong pro-angiogenic capacities. However, a full characterization of the influence of aging on ASC has not been reported yet. Here we've investigated the effect of age on paracrine function, stemness and angiogenic potential of ASC. METHODS ASC were extracted from young and old adult donors. We assessed stromal vascular fraction cell populations repartition, ASC stemness potential, capability to differentiate into mesenchymal lineages as well as their secretome. Angiogenic potential was assessed using a sprouting assay, an indirect co-culture of ASC and dermal microvascular endothelial cells (EC). Total vascular sprout length was measured, and co-culture soluble factors were quantified. Pro-angiogenic factors alone or in combination as well as ASC-conditioned medium (CM) were added to EC to assess sprouting induction. RESULTS Decrease of endothelial cells yield and percentage is observed in cells extracted from adipose tissue of older patients, whereas ASC percentage increased with age. Clonogenic potential of ASC is stable with age. ASC can differentiate into adipocytes, chondrocytes and osteoblasts, and aging does not alter this potential. Among the 25 analytes quantified, high levels of pro-angiogenic factors were found, but none is significantly modulated with age. ASC induce a significantly longer vascular sprouts compared to fibroblasts, and no difference was found between young and old ASC donors on that parameter. Higher concentrations of FGF-2, G-CSF, HGF and IL-8, and lower concentrations of VEGF-C were quantified in EC/ASC co-cultures compared to EC/fibroblasts co-cultures. EC/ASC from young donors secrete higher levels of VEGF-A compared to old ones. Neither soluble factor nor CM without cells are able to induce organized sprouts, highlighting the requirement of cell communication for sprouting. CM produced by ASC supporting development of long vascular sprouts promote sprouting in co-cultures that establish shorter sprouts. CONCLUSION Our results show cells from young and old donors exhibit no difference in all assessed parameters, suggesting all patients could be included in clinical applications. We emphasized the leading role of ASC in angiogenesis, without impairment with age, where secretome is a key but not sufficient actor.
Collapse
Affiliation(s)
- Chloe Trotzier
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France.
| | - Clement Bellanger
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Hakima Abdessadeq
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Philippe Delannoy
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Ali Mojallal
- Department of Plastic, Reconstructive and Aesthetic Surgery, La Croix Rousse Hospital, Bernard Lyon 1 University, Lyon, France
| | - Celine Auxenfans
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
2
|
Uguten M, van der Sluis N, Vriend L, Coert JH, Harmsen MC, van der Lei B, van Dongen JA. Comparing mechanical and enzymatic isolation procedures to isolate adipose-derived stromal vascular fraction: A systematic review. Wound Repair Regen 2024; 32:1008-1021. [PMID: 39444305 PMCID: PMC11584359 DOI: 10.1111/wrr.13228] [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: 07/11/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
The stromal vascular fraction of adipose tissue has gained popularity as regenerative therapy for tissue repair. Both enzymatic and mechanical intraoperative SVF isolation procedures exist. To date, the quest for the preferred isolation procedure persists, due to the absence of standardised yield measurements and a defined clinical threshold. This systematic review is an update of the systematic review published in 2018, where guidelines were proposed to improve and standardise SVF isolation procedures. An elaborate data search in MEDLINE (PubMed), EMBASE (Ovid) and the Cochrane Central Register of Controlled Trials was conducted from September 2016 to date. A total of 26 full-text articles met inclusion criteria, evaluating 33 isolation procedures (11 enzymatic and 22 mechanical). In general, enzymatic and mechanical SVF isolation procedures yield comparable outcomes concerning cell yield (2.3-18.0 × 105 resp. 0.03-26.7 × 105 cells/ml), and cell viability (70%-99% resp. 46%-97.5%), while mechanical procedures are less time consuming (8-20 min vs. 50-210 min) and cost-efficient. However, as most studies used poorly validated outcome measures on SVF characterisation, it still remains unclear which intraoperative SVF isolation method is preferred. Future studies are recommended to implement standardised guidelines to standardise methods and improve comparability between studies.
Collapse
Affiliation(s)
- Mustafa Uguten
- Department of Plastic, Reconstructive and Hand SurgeryMedical Center LeeuwardenLeeuwardenThe Netherlands
- Department of Plastic, Reconstructive and Hand SurgeryUniversity Medical Center Utrecht, University of UtrechtUtrechtThe Netherlands
| | - Nanouk van der Sluis
- Department of SurgeryErasmus University Medical Center, University Medical Center RotterdamRotterdamThe Netherlands
| | - Linda Vriend
- Department of Pathology & Medical BiologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - J. H. Coert
- Department of Plastic, Reconstructive and Hand SurgeryUniversity Medical Center Utrecht, University of UtrechtUtrechtThe Netherlands
| | - Martin C. Harmsen
- Department of Pathology & Medical BiologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - Berend van der Lei
- Department of Plastic SurgeryUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Joris A. van Dongen
- Department of Plastic, Reconstructive and Hand SurgeryUniversity Medical Center Utrecht, University of UtrechtUtrechtThe Netherlands
- Department of Pathology & Medical BiologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
3
|
Diehm YF, Thomé J, Will P, Kotsougiani-Fischer D, Haug VF, Siegwart LC, Kneser U, Fischer S. Stem Cell-Enriched Hybrid Breast Reconstruction Reduces Risk for Capsular Contracture in a Hybrid Breast Model. Plast Reconstr Surg 2023; 152:572-580. [PMID: 36735813 DOI: 10.1097/prs.0000000000010260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hybrid breast reconstruction (HBR) combines silicone implants with fat grafting to improve implant coverage, treating local tissue deficiencies and leading to a more natural breast appearance. Recent data also indicated less capsular contracture after HBR. The authors developed a novel technique and animal model of cell-assisted (CA) HBR to illuminate its effects on capsular contracture. METHODS Animals received silicone implants in a dorsal submuscular pocket. Although animals of the HBR group received fat grafting around the implant without stem cell enrichment, rats of the CA-HBR1 and the CA-HBR2 groups received stem cell-enriched fat grafting with 2 × 10 6 and 4 × 10 6 adipose-derived stem cells immediately after implant insertion. On day 60, animals underwent sonography and elastography imaging and were euthanized, and outcome analysis was performed by means of histology, immunohistochemistry, chemical collagen quantification, and gene expression analysis. RESULTS With this novel technique, long-term survival of adipose-derived stem cells within the implant pocket was demonstrated after 60 days after implant insertion. CA-HBR led to significantly reduced thickness and collagen density of capsular contractures. In addition, CA-HBR resulted in reduced fibrotic responses with less occurrence of collagen type I and transforming growth factor-β in capsule tissue. Moreover, the addition of stem cells suppressed fibrotic and inflammatory responses on a genetic level with significant underexpression of collagen type I and transforming growth factor-β1. CONCLUSIONS With this new technique and animal model, the authors observed a preventive effect on capsular contracture substantiating the basis of clinical outcomes of HBR. The authors propose that the addition of stem cells to HBR might booster its beneficial results. CLINICAL RELEVANCE STATEMENT Stem cell-enriched fat grafting around silicone implants may reduce the risk for capsular contracture after silicone breast implantation. While fat grafting alone already shows beneficial effects, the addition of stem cells to the fat graft can potentiate this effect.
Collapse
Affiliation(s)
- Yannick F Diehm
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Julia Thomé
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Patrick Will
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Dimitra Kotsougiani-Fischer
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
- Private Practice for Plastic and Aesthetic Surgery, Aesthetikon Mannheim and Heidelberg
| | - Valentin F Haug
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Laura C Siegwart
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Ulrich Kneser
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
| | - Sebastian Fischer
- From the Department of Hand, Plastic, and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen; University of Heidelberg
- Private Practice for Plastic and Aesthetic Surgery, Aesthetikon Mannheim and Heidelberg
| |
Collapse
|
4
|
Ciccocioppo R, Guadalajara H, Astori G, Carlino G, García-Olmo D. Misconceptions, hurdles and recommendations regarding the use of mesenchymal stem/stromal cells in perianal Crohn disease. Cytotherapy 2023; 25:230-234. [PMID: 36543715 DOI: 10.1016/j.jcyt.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/14/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy.
| | - Hector Guadalajara
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| | - Giuseppe Astori
- Laboratory of Advanced Cellular Therapies, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Giorgio Carlino
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy
| | - Damián García-Olmo
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| |
Collapse
|
5
|
Vargel İ, Tuncel A, Baysal N, Hartuç-Çevik İ, Korkusuz F. Autologous Adipose-Derived Tissue Stromal Vascular Fraction (AD-tSVF) for Knee Osteoarthritis. Int J Mol Sci 2022; 23:13517. [PMID: 36362308 PMCID: PMC9658499 DOI: 10.3390/ijms232113517] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 07/30/2023] Open
Abstract
Adipose tissue contains adult mesenchymal stem cells that may modulate the metabolism when applied to other tissues. Stromal vascular fraction (SVF) can be isolated from adipose tissue mechanically and/or enzymatically. SVF was recently used to decrease the pain and improve the function of knee osteoarthritis (OA) patients. Primary and/or secondary OA causes inflammation and degeneration in joints, and regenerative approaches that may modify the natural course of the disease are limited. SVF may modulate inflammation and initiate regeneration in joint tissues by initiating a paracrine effect. Chemokines released from SVF may slow down degeneration and stimulate regeneration in joints. In this review, we overviewed articular joint cartilage structures and functions, OA, and macro-, micro-, and nano-fat isolation techniques. Mechanic and enzymatic SVF processing techniques were summarized. Clinical outcomes of adipose tissue derived tissue SVF (AD-tSVF) were evaluated. Medical devices that can mechanically isolate AD-tSVF were listed, and publications referring to such devices were summarized. Recent review manuscripts were also systematically evaluated and included. Transferring adipose tissues and cells has its roots in plastic, reconstructive, and aesthetic surgery. Micro- and nano-fat is also transferred to other organs and tissues to stimulate regeneration as it contains regenerative cells. Minimal manipulation of the adipose tissue is recently preferred to isolate the regenerative cells without disrupting them from their natural environment. The number of patients in the follow-up studies are recently increasing. The duration of follow up is also increasing with favorable outcomes from the short- to mid-term. There are however variations for mean age and the severity of knee OA patients between studies. Positive outcomes are related to the higher number of cells in the AD-tSVF. Repetition of injections and concomitant treatments such as combining the AD-tSVF with platelet rich plasma or hyaluronan are not solidified. Good results were obtained when combined with arthroscopic debridement and micro- or nano-fracture techniques for small-sized cartilage defects. The optimum pressure applied to the tissues and cells during filtration and purification of the AD-tSVF is not specified yet. Quantitative monitoring of articular joint cartilage regeneration by ultrasound, MR, and synovial fluid analysis as well as with second-look arthroscopy could improve our current knowledge on AD-tSVF treatment in knee OA. AD-tSVF isolation techniques and technologies have the potential to improve knee OA treatment. The duration of centrifugation, filtration, washing, and purification should however be standardized. Using gravity-only for isolation and filtration could be a reasonable approach to avoid possible complications of other methodologies.
Collapse
Affiliation(s)
- İbrahim Vargel
- Department of Plastic Reconstructive and Aesthetic Surgery, Medical Faculty, Hacettepe University, Altındag, Ankara 06230, Turkey
| | - Ali Tuncel
- Department of Chemical Engineering, Engineering Faculty, Hacettepe University, Universiteler Mahallesi, Hacettepe Beytepe Campus #31, Çankaya, Ankara 06800, Turkey
| | - Nilsu Baysal
- Medical Faculty, Hacettepe University, Altındag, Ankara 06230, Turkey
| | - İrem Hartuç-Çevik
- Department of Sports Medicine, Medical Faculty, Hacettepe University, Altındag, Ankara 06230, Turkey
| | - Feza Korkusuz
- Department of Sports Medicine, Medical Faculty, Hacettepe University, Altındag, Ankara 06230, Turkey
| |
Collapse
|
6
|
Recent Patents Involving Stromal Vascular Fraction. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00283-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
7
|
Boada-Pladellorens A, Avellanet M, Pages-Bolibar E, Veiga A. Stromal vascular fraction therapy for knee osteoarthritis: a systematic review. Ther Adv Musculoskelet Dis 2022; 14:1759720X221117879. [PMID: 35991523 PMCID: PMC9386815 DOI: 10.1177/1759720x221117879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/19/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Regenerative cell therapies, such as adipose-derived stromal vascular fraction (SVF), have been postulated as potential treatments for knee osteoarthritis (KOA). Objectives: To assess the efficacy and safety of SVF treatment against placebo and other standard therapies for treating KOA in adult patients. Design: A systematic review. Data sources and methods: We searched the following databases: MEDLINE via PubMed, Epistemonikos, PEDro, DynaMed, TripDatabase, Elsevier via Clinicalkey and Cochrane Controlled Trials Register. We included prospective interventional studies where treatment with SVF in adults with KOA was compared against placebo or other standard therapies, and results were objectively measured with at least one widely recognised osteoarthritis scale. Results: Among 266 studies published until May 2021, nine met our inclusion criteria. A total of 239 patients (274 knees) were included in our study. The follow-up ranged from 6 to 24 months. Six studies had a control group (only one being placebo). All studies showed that SVF improved pain and functionality measured, in most cases, with the visual analogue scale and the Western Ontario and McMaster Universities Osteoarthritis Index. In addition, five studies reported an improvement in anatomical structures, as detected in MR images. However, the number of cells contained in SVF varied substantially between different studies, which could induce a comparison bias. Conclusion: Although based on a small number of dissimilar studies, SVF was considered a safe treatment for KOA and could be promising in terms of pain, functionality and anatomical structure improvement. However, SVF products need to be standardised, the number of cells homogenised and the use of concomitant treatments reduced to establish proper comparisons. Registration: PROSPERO registration number: CRD42021284187.
Collapse
Affiliation(s)
- Anna Boada-Pladellorens
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Carrer dels Escalls, AD700 Escaldes-Engordany, Andorra
| | - Mercè Avellanet
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Escaldes-Engordany, Andorra
| | - Esther Pages-Bolibar
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Escaldes-Engordany, Andorra
| | - Anna Veiga
- Barcelona Stem Cell Bank, Regenerative Medicine Programme, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, Barcelona, Spain
| |
Collapse
|
8
|
Verdura V, Guastafierro A, Di Pace B, Faenza M, Nicoletti GF, Rubino C. Optimizing Fat Grafting Using a Hydraulic System Technique for Fat Processing: A Time and Cost Analysis. Arch Plast Surg 2022; 49:266-274. [PMID: 35832664 PMCID: PMC9045492 DOI: 10.1055/s-0042-1744361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background
Many authors have researched ways to optimize fat grafting by looking for a technique that offers safe and long-term fat survival rate. To date, there is no standardized protocol. We designed a “hydraulic system technique” optimizing the relationship among the quantity of injected fat, operative time, and material cost to establish fat volume cutoffs for a single procedure.
Methods
Thirty-six patients underwent fat grafting surgery and were organized into three groups according to material used: standard, “1-track,” and “2-tracks” systems. The amount of harvested and grafted fat as well as material used for each procedure was collected. Operating times were recorded and statistical analysis was performed to establish the relationship with the amount of treated fat.
Results
In 15 cases the standard system was used (mean treated fat 72 [30–100] mL, mean cost 4.23 ± 0.27 euros), in 11 cases the “1-track” system (mean treated fat 183.3 [120–280] mL, mean cost 7.63 ± 0.6 euros), and in 10 cases the “2-tracks” one (mean treated fat 311[220–550] mL, mean cost 12.47 ± 1 euros). The mean time difference between the standard system and the “1-track” system is statistically significant starting from three fat syringes (90 mL) in 17.66 versus 6.87 minutes. The difference between the “1-track” system and “2-tracks” system becomes statistically significant from 240 mL of fat in 15 minutes (“1-track”) versus 9.3 minutes for the “2-tracks” system.
Conclusion
Data analysis would indicate the use of the standard system, “1-track,” and “2-tracks” to treat an amount of fat < 90 mL of fat, 90 ÷ 240 mL of fat, and ≥ 240 mL of fat, respectively.
Collapse
Affiliation(s)
- Vincenzo Verdura
- Multidisciplinary Department of Medical Surgical and Dental Specialties, Plastic Surgery Unit, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Antonio Guastafierro
- Multidisciplinary Department of Medical Surgical and Dental Specialties, Plastic Surgery Unit, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Bruno Di Pace
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana,” PhD School of Translational Medicine of Development and Active Aging, University of Salerno, Salerno, Italy
- Department of Plastic and Reconstructive Surgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Anglia Ruskin University School of Medicine, Cambridge and Chelmsford, United Kingdom
| | - Mario Faenza
- Multidisciplinary Department of Medical Surgical and Dental Specialties, Plastic Surgery Unit, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical Surgical and Dental Specialties, Plastic Surgery Unit, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Corrado Rubino
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
- Plastic Surgery Unit, Department of Oncology and Haematology, University Hospital Trust of Sassari, Sassari, Italy
| |
Collapse
|
9
|
Immunomodulation of Skin Repair: Cell-Based Therapeutic Strategies for Skin Replacement (A Comprehensive Review). Biomedicines 2022; 10:biomedicines10010118. [PMID: 35052797 PMCID: PMC8773777 DOI: 10.3390/biomedicines10010118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system has a crucial role in skin wound healing and the application of specific cell-laden immunomodulating biomaterials emerged as a possible treatment option to drive skin tissue regeneration. Cell-laden tissue-engineered skin substitutes have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. In particular, mesenchymal stem cells with their immunomodulatory properties can create a specific immune microenvironment to reduce inflammation, scarring, and support skin regeneration. This review presents an overview of current wound care techniques including skin tissue engineering and biomaterials as a novel and promising approach. We highlight the plasticity and different roles of immune cells, in particular macrophages during various stages of skin wound healing. These aspects are pivotal to promote the regeneration of nonhealing wounds such as ulcers in diabetic patients. We believe that a better understanding of the intrinsic immunomodulatory features of stem cells in implantable skin substitutes will lead to new translational opportunities. This, in turn, will improve skin tissue engineering and regenerative medicine applications.
Collapse
|
10
|
Ragni E, Viganò M, De Luca P, Pedrini E, de Girolamo L. Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue. ORTHOBIOLOGICS 2022:47-61. [DOI: 10.1007/978-3-030-84744-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
11
|
Sharma S, Muthu S, Jeyaraman M, Ranjan R, Jha SK. Translational products of adipose tissue-derived mesenchymal stem cells: Bench to bedside applications. World J Stem Cells 2021; 13:1360-1381. [PMID: 34786149 PMCID: PMC8567449 DOI: 10.4252/wjsc.v13.i10.1360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
With developments in the field of tissue engineering and regenerative medicine, the use of biological products for the treatment of various disorders has come into the limelight among researchers and clinicians. Among all the available biological tissues, research and exploration of adipose tissue have become more robust. Adipose tissue engineering aims to develop by-products and their substitutes for their regenerative and immunomodulatory potential. The use of biodegradable scaffolds along with adipose tissue products has a major role in cellular growth, proliferation, and differentiation. Adipose tissue, apart from being the powerhouse of energy storage, also functions as the largest endocrine organ, with the release of various adipokines. The progenitor cells among the heterogeneous population in the adipose tissue are of paramount importance as they determine the capacity of regeneration of these tissues. The results of adipose-derived stem-cell assisted fat grafting to provide numerous growth factors and adipokines that improve vasculogenesis, fat graft integration, and survival within the recipient tissue and promote the regeneration of tissue are promising. Adipose tissue gives rise to various by-products upon processing. This article highlights the significance and the usage of various adipose tissue by-products, their individual characteristics, and their clinical applications.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
| | - Sathish Muthu
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul, Tamil Nadu 624304, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| |
Collapse
|
12
|
Update on the Basic Science Concepts and Applications of Adipose-Derived Stem Cells in Hand and Craniofacial Surgery. Plast Reconstr Surg 2021; 148:475e-486e. [PMID: 34432707 DOI: 10.1097/prs.0000000000008279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
SUMMARY Adipose-derived stem cell therapy offers plastic surgeons a novel treatment alternative for conditions with few therapeutic options. Adipose-derived stem cells are a promising treatment because of their broad differentiation potential, capacity for self-renewal, and ease of isolation. Over the past decade, plastic surgeons have attempted to harness adipose-derived stem cells' unique cellular characteristics to improve the survival of traditional fat grafting procedures, a process known as cell-assisted lipotransfer. However, the full implications of cell-assisted lipotransfer in clinical practice remain incompletely understood, stressing the urgent need to assess the scientific evidence supporting adipose-derived stem cell-based interventions. Furthermore, with the strict regulatory climate surrounding tissue explantation therapies, reviewing the safety and efficacy of these treatments will clarify their regulatory viability moving forward. In this report, the authors provide a comprehensive, up-to-date appraisal of best evidence-based practices supporting adipose-derived stem cell-derived therapies, highlighting the known mechanisms behind current clinical applications in tissue engineering and regenerative medicine specific to plastic and reconstructive surgery. The authors outline best practices for the harvest and isolation of adipose-derived stem cells and discuss why procedure standardization will elucidate the scientific bases for their broad use. Finally, the authors discuss challenges posed by U.S. Food and Drug Administration oversight of these cell-based therapies and examine the role of adipose-derived stem cell-based applications in the future of plastic surgery.
Collapse
|
13
|
Ude CC, Shah S, Ogueri KS, Nair LS, Laurencin CT. Stromal Vascular Fraction for Osteoarthritis of the Knee Regenerative Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 8:210-224. [PMID: 35958164 PMCID: PMC9365234 DOI: 10.1007/s40883-021-00226-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Purpose The knee joint is prone to osteoarthritis (OA) due to its anatomical position, and several reports have implicated the imbalance between catabolic and anabolic processes within the joint as the main culprit, thus leading to investigations towards attenuation of these inflammatory signals for OA treatment. In this review, we have explored clinical evidence supporting the use of stromal vascular fraction (SVF), known for its anti-inflammatory characteristics for the treatment of OA. Methods Searches were made on PubMed, PMC, and Google Scholar with the keywords “adipose fraction knee regeneration, and stromal vascular fraction knee regeneration, and limiting searches within 2017–2020. Results Frequently found interventions include cultured adipose-derived stem cells (ADSCs), SVF, and the micronized/microfragmented adipose tissue-stromal vascular fraction (MAT-SVF). Clinical data reported that joints treated with SVF provided a better quality of life to patients. Currently, MAT-SVF obtained and administered at the point of care is approved by the Food and Drug Administration (FDA), but more studies including manufacturing validation, safety, and proof of pharmacological activity are needed for SVF. The mechanism of action of MAT-SVF is also not fully understood. However, the current hypothesis indicates a direct adherence and integration with the degenerative host tissue, and/or trophic effects resulting from the secretome of constituent cells. Conclusion Our review of the literature on stromal vascular fraction and related therapy use has found evidence of efficacy in results. More research and clinical patient follow-up are needed to determine the proper place of these therapies in the treatment of osteoarthritis of the knee. Lay Summary Reports have implicated the increased inflammatory proteins within the joints as the main cause of osteoarthritis (OA). This has attracted interest towards addressing these inflammatory proteins as a way of treatment for OA. The concentrated cell-packed portion of the adipose product stromal vascular fraction (SVF) from liposuction or other methods possesses anti-inflammatory effects and has been acclaimed to heal OA. Thus, we searched for clinical evidence supporting their use, for OA treatment through examining the literature. Data from various hospitals support that joints treated with SVF provided a better quality of life to patients. Currently, there is at least one version of these products that are obtained and given back to patients during a single clinic visit, approved by the FDA.
Collapse
Affiliation(s)
- Chinedu C. Ude
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Farmington, CT, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Shiv Shah
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Farmington, CT, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
| | - Kenneth S. Ogueri
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Farmington, CT, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, USA
| | - Lakshmi S. Nair
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Farmington, CT, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Cato T. Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Farmington, CT, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, USA
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
14
|
François P, Rusconi G, Arnaud L, Mariotta L, Giraudo L, Minonzio G, Veran J, Bertrand B, Dumoulin C, Grimaud F, Lyonnet L, Casanova D, Giverne C, Cras A, Magalon G, Dignat-George F, Sabatier F, Magalon J, Soldati G. Inter-center comparison of good manufacturing practices-compliant stromal vascular fraction and proposal for release acceptance criteria: a review of 364 productions. Stem Cell Res Ther 2021; 12:373. [PMID: 34210363 PMCID: PMC8252207 DOI: 10.1186/s13287-021-02445-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
Background Even though the manufacturing processes of the stromal vascular fraction for clinical use are performed in compliance with the good manufacturing practices applying to advanced therapy medicinal products, specifications related to stromal vascular fraction quality remain poorly defined. We analyzed stromal vascular fraction clinical batches from two independent good manufacturing practices-compliant manufacturing facilities, the Swiss Stem Cell Foundation (SSCF) and Marseille University Hospitals (AP-HM), with the goal of defining appropriate and harmonized release acceptance criteria. Methods This retrospective analysis reviewed the biological characteristics of 364 batches of clinical-grade stromal vascular fraction. Collected data included cell viability, recovery yield, cell subset distribution of stromal vascular fraction, and microbiological quality. Results Stromal vascular fraction from SSCF cohort demonstrated a higher viability (89.33% ± 4.30%) and recovery yield (2.54 × 105 ± 1.22 × 105 viable nucleated cells (VNCs) per mL of adipose tissue) than stromal vascular fraction from AP-HM (84.20% ± 5.96% and 2.25 × 105 ± 1.11 × 105 VNCs per mL). AP-HM batches were significantly less contaminated (95.71% of sterile batches versus 74.15% for SSCF batches). The cell subset distribution was significantly different (higher proportion of endothelial cells and lower proportion of leukocytes and pericytes in SSCF cohort). Conclusions Both centers agreed that a good manufacturing practices-compliant stromal vascular fraction batch should exert a viability equal or superior to 80%, a minimum recovery yield of 1.50 × 105 VNCs per mL of adipose tissue, a proportion of adipose-derived stromal cells at least equal to 20%, and a proportion of leukocytes under 50%. In addition, a multiparameter gating strategy for stromal vascular fraction analysis is proposed. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02445-z.
Collapse
Affiliation(s)
- Pauline François
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Giulio Rusconi
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland.,Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Laurent Arnaud
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Luca Mariotta
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| | - Laurent Giraudo
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Greta Minonzio
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| | - Julie Veran
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Baptiste Bertrand
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Chloé Dumoulin
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Fanny Grimaud
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Luc Lyonnet
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Dominique Casanova
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Camille Giverne
- Normandie Univ, UNIROUEN, INSERM, U1234, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Audrey Cras
- Assistance Publique-Hôpitaux de Paris, Saint-Louis Hospital, Cell Therapy Unit, Cord blood Bank and CIC-BT501, Paris, France
| | | | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Remedex, Marseille, France
| | - Jeremy Magalon
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France. .,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France. .,Remedex, Marseille, France.
| | - Gianni Soldati
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| |
Collapse
|
15
|
Jayaraman P, Lim R, Ng J, Vemuri MC. Acceleration of Translational Mesenchymal Stromal Cell Therapy Through Consistent Quality GMP Manufacturing. Front Cell Dev Biol 2021; 9:648472. [PMID: 33928083 PMCID: PMC8076909 DOI: 10.3389/fcell.2021.648472] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stromal cell (hMSC) therapy has been gaining immense interest in regenerative medicine and quite recently for its immunomodulatory properties in COVID-19 treatment. Currently, the use of hMSCs for various diseases is being investigated in >900 clinical trials. Despite the huge effort, setting up consistent and robust scalable manufacturing to meet regulatory compliance across various global regions remains a nagging challenge. This is in part due to a lack of definitive consensus for quality control checkpoint assays starting from cell isolation to expansion and final release criterion of clinical grade hMSCs. In this review, we highlight the bottlenecks associated with hMSC-based therapies and propose solutions for consistent GMP manufacturing of hMSCs starting from raw materials selection, closed and modular systems of manufacturing, characterization, functional testing, quality control, and safety testing for release criteria. We also discuss the standard regulatory compliances adopted by current clinical trials to broaden our view on the expectations across different jurisdictions worldwide.
Collapse
Affiliation(s)
| | - Ryan Lim
- Thermo Fisher Scientific, Singapore, Singapore
| | | | | |
Collapse
|
16
|
Al-Ghadban S, Bunnell BA. Adipose Tissue-Derived Stem Cells: Immunomodulatory Effects and Therapeutic Potential. Physiology (Bethesda) 2021; 35:125-133. [PMID: 32027561 DOI: 10.1152/physiol.00021.2019] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) can self-renew and differentiate along multiple cell lineages. ASCs are also potently anti-inflammatory due to their inherent ability to regulate the immune system by secreting anti-inflammatory cytokines and growth factors that play a crucial role in the pathology of many diseases, including multiple sclerosis, diabetes mellitus, Crohn's, SLE, and graft-versus-host disease. The immunomodulatory effects and mechanisms of action of ASCs on pathological conditions are reviewed here.
Collapse
Affiliation(s)
- Sara Al-Ghadban
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bruce A Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
17
|
Mazini L, Ezzoubi M, Malka G. Overview of current adipose-derived stem cell (ADSCs) processing involved in therapeutic advancements: flow chart and regulation updates before and after COVID-19. Stem Cell Res Ther 2021; 12:1. [PMID: 33397467 PMCID: PMC7781178 DOI: 10.1186/s13287-020-02006-w] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) have raised big interest in therapeutic applications in regenerative medicine and appear to fulfill the criteria for a successful cell therapy. Their low immunogenicity and their ability to self-renew, to differentiate into different tissue-specific progenitors, to migrate into damaged sites, and to act through autocrine and paracrine pathways have been altogether testified as the main mechanisms whereby cell repair and regeneration occur. The absence of standardization protocols in cell management within laboratories or facilities added to the new technologies improved at patient's bedside and the discrepancies in cell outcomes and engraftment increase the limitations on their widespread use by balancing their real benefit versus the patient safety and security. Also, comparisons across pooled patients are particularly difficult in the fact that multiple medical devices are used and there is absence of harmonized assessment assays despite meeting regulations agencies and efficient GMP protocols. Moreover, the emergence of the COVID-19 breakdown added to the complexity of implementing standardization. Cell- and tissue-based therapies are completely dependent on the biological manifestations and parameters associated to and induced by this virus where the scope is still unknown. The initial flow chart identified for stem cell therapies should be reformulated and updated to overcome patient infection and avoid significant variability, thus enabling more patient safety and therapeutic efficiency. The aim of this work is to highlight the major guidelines and differences in ADSC processing meeting the current good manufacturing practices (cGMP) and the cellular therapy-related policies. Specific insights on standardization of ADSCs proceeding at different check points are also presented as a setup for the cord blood and bone marrow.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Center of Biological and Medical Sciences CIAM, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, 43150 Ben Guerir, Morocco
| | - Mohamed Ezzoubi
- Centre des Brûlés et chirurgie réparatrice, Centre Hospitalier Universitaire Ibn Rochd Casablanca, Faculté de Médecine et de Pharmacie Casablanca, Casablanca, Morocco
| | - Gabriel Malka
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Center of Biological and Medical Sciences CIAM, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, 43150 Ben Guerir, Morocco
| |
Collapse
|
18
|
Busato A, De Francesco F, Biswas R, Mannucci S, Conti G, Fracasso G, Conti A, Riccio V, Riccio M, Sbarbati A. Simple and Rapid Non-Enzymatic Procedure Allows the Isolation of Structurally Preserved Connective Tissue Micro-Fragments Enriched with SVF. Cells 2020; 10:cells10010036. [PMID: 33383682 PMCID: PMC7824313 DOI: 10.3390/cells10010036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/20/2020] [Accepted: 12/25/2020] [Indexed: 12/25/2022] Open
Abstract
The stromal vascular fraction (SVF) consists of a heterogeneous population of stem and stromal cells, generally obtained from adipose tissue by enzymatic digestion. For human cell-based therapies, mechanical process methods to obtain SVF represent an advantageous approach because they have fewer regulatory restrictions for their clinical use. The aim of this study was to characterize a novel commercial system for obtaining SVF from adipose tissue by a mechanical approach without substantial manipulations. Lipoaspirate samples collected from 27 informed patients were processed by a simple and fast mechanical system (by means of Hy-Tissue SVF). The Hy-Tissue SVF product contained a free cell fraction and micro-fragments of stromal connective tissue. The enzymatic digestion of the micro-fragments increased the yield of free cells (3.2 times) and CFU-F (2.4 times). Additionally, 10% of free cells from SVF were positive for CD34+, suggesting the presence of endothelial cells, pericytes, and potential adipose-derived stem cells (ADSC). Moreover, the SVF cells were able to proliferate and differentiate in vitro toward adipocytes, osteocytes, and chondrocytes. The immunophenotypic analysis of expanded cells showed positivity for typical mesenchymal stem cell markers. The Hy-Tissue SVF system allows the isolation of stromal vascular fraction, making this product of potential interest in regenerative medicine.
Collapse
Affiliation(s)
- Alice Busato
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU “Ospedali Riuniti”, 60126 Ancona, Italy; (F.D.F.); (M.R.)
- Correspondence: ; Tel.: +39-071-596-3945
| | - Reetuparna Biswas
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
| | - Silvia Mannucci
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
| | - Giamaica Conti
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
| | - Giulio Fracasso
- Department of Medicine, Section of Immunology, University of Verona, 37135 Verona, Italy;
| | - Anita Conti
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
| | - Valentina Riccio
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy;
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU “Ospedali Riuniti”, 60126 Ancona, Italy; (F.D.F.); (M.R.)
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, 61025 Montelabbate, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37135 Verona, Italy; (A.B.); (R.B.); (S.M.); (G.C.); (A.C.); (A.S.)
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, 61025 Montelabbate, Italy
| |
Collapse
|
19
|
Development and Validation of a Fully GMP-Compliant Process for Manufacturing Stromal Vascular Fraction: A Cost-Effective Alternative to Automated Methods. Cells 2020; 9:cells9102158. [PMID: 32987708 PMCID: PMC7598595 DOI: 10.3390/cells9102158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
The therapeutic use of adipose-derived stromal vascular fraction (SVF) is expanding in multiple pathologies. Various processes have been proposed for manufacturing SVF but they must be revisited based on advanced therapy medicinal product (ATMP) regulations. We report here the development and validation of a fully good manufacturing practices (GMP)-compliant protocol for the isolation of SVF. Adipose tissue was collected from healthy volunteers undergoing lipoaspiration. The optimal conditions of collagenase digestion and washing were determined based on measurements of SVF cell viability, yield recovery, and cell subset distribution. Comparability of the SVF obtained using the newly developed manufacturing process (n = 6) and the Celution-based automated method (n = 33), used as a reference, was established using inter-donor analyses. Characteristics of SVF (n = 5) generated using both manufacturing protocols were analyzed for an intra-donor comparison. In addition, these comparisons also included the determination of colony-forming unit fibroblast frequency, in vitro angiogenic activity, and in vivo regenerative effects in a mouse ischemic cutaneous wound model. We successfully developed a process for the generation of SVF presenting higher cell viability and yield recovery compared to the Celution device-based protocol. Characteristics of the SVF including phenotype, capacity for angiogenesis, and wound-healing promotion attested to the comparability of the two manufacturing processes. We validated an optimized non-automated process that should allow for a GMP-compliant, more affordable, and reduced-cost strategy to exploit the potential of SVF-based regenerative therapies.
Collapse
|
20
|
Guillaume O, Pérez-Köhler B, Schädl B, Keibl C, Saxenhuber N, Heimel P, Priglinger E, Wolbank S, Redl H, Petter-Puchner A, Fortelny R. Stromal vascular fraction cells as biologic coating of mesh for hernia repair. Hernia 2020; 24:1233-1243. [PMID: 32096088 PMCID: PMC7701131 DOI: 10.1007/s10029-020-02135-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/29/2020] [Indexed: 12/19/2022]
Abstract
Background The interest in non-manipulated cells originating from adipose tissue has raised tremendously in the field of tissue engineering and regenerative medicine. The resulting stromal vascular fraction (SVF) cells have been successfully used in numerous clinical applications. The aim of this experimental work is, first to combine a macroporous synthetic mesh with SVF isolated using a mechanical disruption process, and to assess the effect of those cells on the early healing phase of hernia. Methods Human SVF cells combined with fibrin were used to coat commercial titanized polypropylene meshes. In vitro, viability and growth of the SVF cells were assessed using live/dead staining and scanning electron microscopy. The influence of SVF cells on abdominal wall hernia healing was conducted on immunodeficient rats, with a focus on short-term vascularization and fibrogenesis. Results Macroporous meshes were easily coated with SVF using a fibrin gel as temporary carrier. The in vitro experiments showed that the whole process including the isolation of human SVF cells and their coating on PP meshes did not impact on the SVF cells’ viability and on their capacity to attach and to proliferate. In vivo, the SVF cells were well tolerated by the animals, and coating mesh with SVF resulted in a decrease degree of vascularity compared to control group at day 21. Conclusions The utilization of SVF-coated mesh influences the level of angiogenesis during the early onset of tissue healing. Further long-term animal experiments are needed to confirm that this effect correlates with a more robust mesh integration compared to non-SVF-coated mesh.
Collapse
Affiliation(s)
- O Guillaume
- 3D Printing and Biofabrication Group, Institute of Materials Science and Technology, TU Wien, Vienna, Austria. .,Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - B Pérez-Köhler
- Department of Medicine and Medical Specialties, University of Alcalá, Madrid, Spain.,Biomedical Networking Research Centre On Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - B Schädl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - C Keibl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - N Saxenhuber
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - P Heimel
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - E Priglinger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - S Wolbank
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - H Redl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - A Petter-Puchner
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of General, Visceral and Oncologic Surgery, Wilhelminenspital, Vienna, Austria
| | - R Fortelny
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of General, Visceral and Oncologic Surgery, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
21
|
Hong KY, Kim IK, Park SO, Jin US, Chang H. Reply: Systemic Administration of Adipose-Derived Stromal Cells Concurrent with Fat Grafting. Plast Reconstr Surg 2019; 144:1115e-1116e. [PMID: 31764690 DOI: 10.1097/prs.0000000000006218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Dongguk University Medical Center, Goyang, Republic of Korea
| | - Il-Kug Kim
- Department of Plastic and Reconstructive Surgery, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Seong Oh Park
- Department of Plastic and Reconstructive Surgery, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Ung Sik Jin
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hak Chang
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Kunisch E, Gunnella F, Wagner S, Dees F, Maenz S, Bossert J, Jandt KD, Kinne RW. The poly (l-lactid-co-glycolide; PLGA) fiber component of brushite-forming calcium phosphate cement induces the osteogenic differentiation of human adipose tissue-derived stem cells. ACTA ACUST UNITED AC 2019; 14:055012. [PMID: 31465298 DOI: 10.1088/1748-605x/ab3544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A brushite-forming calcium phosphate cement (CPC) was mechanically stabilized by addition of poly (l-lactid-co-glycolide; PLGA) fibers (≤10% w/w). It proved highly biocompatible and its fiber component enhanced bone formation in a sheep lumbar vertebroplasty model. However, possible effects on the osteogenic differentiation of resident mesenchymal stem cells (MSCs) remained unexplored. The present study used a novel approach, simultaneously analyzing the influence of a solid CPC scaffold and its relatively low PLGA proportion (a mimicry of natural bone) on osteogenic, chondrogenic, and adipogenic differentiation, as well as the pluripotency of human adipose tissue-derived mesenchymal stem cells (hASCs). hASCs were cultured on CPC discs with/without PLGA fibers (5% and 10%) in the absence of osteogenic medium for 3, 7, and 14 d. Gene expression of osteogenic markers (Runx2, osterix, alkaline phosphatase, collagen I, osteonectin, osteopontin, osteocalcin), chondrogenic markers (collagen II, Sox9, aggrecan), adipogenic markers (PPARG, Leptin, and FABP4), and pluripotency markers (Nanog, Tert, Rex) was analyzed by RT-PCR. The ability of hASCs to synthesize alkaline phosphatase was also evaluated. Cell number and viability were determined by fluorescein diacetate/propidium iodide staining. Compared to pure CPC, cultivation of hASCs on fiber-reinforced CPC transiently induced the gene expression of Runx2 and osterix (day 3), and long-lastingly augmented the expression of alkaline phosphatase (and its enzyme activity), collagen I, and osteonectin (until day 14). In contrast, augmented expression of all chondrogenic, adipogenic, and pluripotency markers was limited to day 3, followed by significant downregulation. Cultivation of hASCs on fiber-reinforced CPC reduced the cell number, but not the proportion of viable cells (viability > 95%). The PLGA component of fiber-reinforced, brushite-forming CPC supports long-lasting osteogenic differentiation of hASCs, whereas chondrogenesis, adipogenesis, and pluripotency are initially augmented, but subsequently suppressed. In view of parallel animal results, PLGA fibers may represent an interesting clinical target for future improvement of CPC- based bone regeneration.
Collapse
Affiliation(s)
- Elke Kunisch
- Experimental Rheumatology Unit, Department of Orthopedics, Jena University Hospital, Waldkliniken Eisenberg GmbH, Eisenberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Chu DT, Nguyen Thi Phuong T, Tien NLB, Tran DK, Minh LB, Thanh VV, Gia Anh P, Pham VH, Thi Nga V. Adipose Tissue Stem Cells for Therapy: An Update on the Progress of Isolation, Culture, Storage, and Clinical Application. J Clin Med 2019; 8:E917. [PMID: 31247996 PMCID: PMC6678927 DOI: 10.3390/jcm8070917] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/10/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue stem cells (ASCs), known as multipotent stem cells, are most commonly used in the clinical applications in recent years. Adipose tissues (AT) have the advantage in the harvesting, isolation, and expansion of ASCs, especially an abundant amount of stem cells compared to bone marrow. ASCs can be found in stromal vascular fractions (SVF) which are easily obtained from the dissociation of adipose tissue. Both SVFs and culture-expanded ASCs exhibit the stem cell characteristics such as differentiation into multiple cell types, regeneration, and immune regulators. Therefore, SVFs and ASCs have been researched to evaluate the safety and benefits for human use. In fact, the number of clinical trials on ASCs is going to increase by years; however, most trials are in phase I and II, and lack phase III and IV. This systemic review highlights and updates the process of the harvesting, characteristics, isolation, culture, storage, and application of ASCs, as well as provides further directions on the therapeutic use of ASCs.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam.
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam
| | - Le Bui Minh
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh St., Ward 13, District 4, Ho Chi Minh City 700000, Vietnam
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Pham Gia Anh
- Oncology Department, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Vu Thi Nga
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam.
| |
Collapse
|
24
|
Characterization of Different Sources of Human MSCs Expanded in Serum-Free Conditions with Quantification of Chondrogenic Induction in 3D. Stem Cells Int 2019; 2019:2186728. [PMID: 31320905 PMCID: PMC6610765 DOI: 10.1155/2019/2186728] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/11/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent alternative candidates to chondrocytes for cartilage engineering. However, it remains difficult to identify the ideal source of MSCs for cartilage repair since conditions supporting chondrogenic induction are diverse among published works. In this study, we characterized and evaluated the chondrogenic potential of MSCs from bone marrow (BM), Wharton's jelly (WJ), dental pulp (DP), and adipose tissue (AT) isolated and cultivated under serum-free conditions. BM-, WJ-, DP-, and AT-MSCs did not differ in terms of viability, clonogenicity, and proliferation. By an extensive polychromatic flow cytometry analysis, we found notable differences in markers of the osteochondrogenic lineage between the 4 MSC sources. We then evaluated their chondrogenic potential in a micromass culture model, and only BM-MSCs showed chondrogenic conversion. This chondrogenic differentiation was specifically ascertained by the production of procollagen IIB, the only type II collagen isoform synthesized by well-differentiated chondrocytes. As a pilot study toward cartilage engineering, we encapsulated BM-MSCs in hydrogel and developed an original method to evaluate their chondrogenic conversion by flow cytometry analysis, after release of the cells from the hydrogel. This allowed the simultaneous quantification of procollagen IIB and α10, a subunit of a type II collagen receptor crucial for proper cartilage development. This work represents the first comparison of detailed immunophenotypic analysis and chondrogenic differentiation potential of human BM-, WJ-, DP-, and AT-MSCs performed under the same serum-free conditions, from their isolation to their induction. Our study, achieved in conditions compliant with clinical applications, highlights that BM-MSCs are good candidates for cartilage engineering.
Collapse
|
25
|
Characterization and Optimization of the Seeding Process of Adipose Stem Cells on the Polycaprolactone Scaffolds. Stem Cells Int 2019; 2019:1201927. [PMID: 30915123 PMCID: PMC6402208 DOI: 10.1155/2019/1201927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/24/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
The purpose of the current study was to evaluate the usefulness of adipose-derived stem cells (ASCs) for bone injury therapy. Lipoaspirates were collected from the abdomen regions of 17 healthy female donors (mean age 49 ± 6 years) using Coleman technique or Body-jet liposuction. In the present study, the primary objective was the in vitro characteristics of human ASCs. The secondary objective was the optimization of the cell seeding process on 3D-printed scaffolds using polycaprolactone (PCL) or polycaprolactone covered with tricalcium phosphate (PCL + 5% TCP). Biological evaluation of human ASC showed high efficiency of isolation obtaining a satisfying amount of homogeneous cell populations. Results suggest that ASCs can be cultured in vitro for a long time without impairing their proliferative capacity. Growth kinetics shows that the highest number of cells can be achieved in passage 5 and after the 16th passage; there is a significant decrease of cell numbers and their proliferative potential. The percentage of colony forming units from the adipose stem cells is 8% ± 0.63% (p < 0.05). It was observed that the accumulation of calcium phosphate in the cells in vitro, marked with Alizarin Red S, was increased along with the next passage. Analysis of key parameters critically related to the cell seeding process shows that volume of cell suspension and propagation time greatly improve the efficiency of seeding both in PCL and PCL + 5% TCP scaffolds. The cell seeding efficiency did differ significantly between scaffold materials and cell seeding methods (p < 0.001). Increased seeding efficiency was observed when using the saturation of cell suspension into scaffolds with additional incubation. Alkaline phosphatase level production in PCL + 5% TCP scaffold was better than in PCL-only scaffold. The study results can be used for the optimization of the seeding process and quantification methods determining the successful implementation of the preclinical model study in the future tissue engineering strategies.
Collapse
|
26
|
Parsons AM, Ciombor DM, Liu PY, Darling EM. Regenerative Potential and Inflammation-Induced Secretion Profile of Human Adipose-Derived Stromal Vascular Cells Are Influenced by Donor Variability and Prior Breast Cancer Diagnosis. Stem Cell Rev Rep 2018; 14:546-557. [PMID: 29663271 DOI: 10.1007/s12015-018-9813-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adipose tissue contains a heterogeneous population of stromal vascular fraction (SVF) cells that work synergistically with resident cell types to enhance tissue healing. Ease of access and processing paired with therapeutic promise make SVF cells an attractive option for autologous applications in regenerative medicine. However, inherent variability in SVF cell therapeutic potential from one patient to another hinders prognosis determination for any one person. This study investigated the regenerative properties and inflammation responses of thirteen, medically diverse human donors. Using non-expanded primary lipoaspirate samples, SVF cells were assessed for robustness of several parameters integral to tissue regeneration, including yield, viability, self-renewal capacity, proliferation, differentiation potential, and immunomodulatory cytokine secretion. Each parameter was selected either for its role in regenerative potential, defined here as the ability to heal tissues through stem cell repopulation and subsequent multipotent differentiation, or for its potential role in wound healing through trophic immunomodulatory activity. These data were then analyzed for consistent and predictable patterns between and across measurements, while also investigating the influence of the donors' relevant medical histories, particularly if the donor was in remission following breast cancer treatment. Analyses identified positive correlations among the expression of three cytokines: interleukin (IL)-6, IL-8, and monocyte chemoattractant protein (MCP)-1. The expression of these cytokines also positively related to self-renewal capacity. These results are potentially relevant for establishing expectations in both preclinical experiments and targeted clinical treatment strategies that use stem cells from patients with diverse medical histories.
Collapse
Affiliation(s)
- Adrienne M Parsons
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, 175 Meeting Street, Box G-B397, Providence, RI, 02912, USA
| | - Deborah M Ciombor
- Department of Plastic and Reconstructive Surgery, Brown University, Providence, RI, USA
- Center for Biomedical Engineering, Brown University, Providence, RI, USA
| | - Paul Y Liu
- Department of Plastic and Reconstructive Surgery, Brown University, Providence, RI, USA
| | - Eric M Darling
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, 175 Meeting Street, Box G-B397, Providence, RI, 02912, USA.
- Center for Biomedical Engineering, Brown University, Providence, RI, USA.
- School of Engineering, Brown University, Providence, RI, USA.
- Department of Orthopaedics, Brown University, Providence, RI, USA.
| |
Collapse
|
27
|
Haskett DG, Saleh KS, Lorentz KL, Josowitz AD, Luketich SK, Weinbaum JS, Kokai LE, D'Amore A, Marra KG, Rubin JP, Wagner WR, Vorp DA. An exploratory study on the preparation and evaluation of a "same-day" adipose stem cell-based tissue-engineered vascular graft. J Thorac Cardiovasc Surg 2018; 156:1814-1822.e3. [PMID: 30057192 DOI: 10.1016/j.jtcvs.2018.05.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/17/2018] [Accepted: 05/31/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Tissue-engineered vascular grafts containing adipose-derived mesenchymal stem cells offer an alternative to small-diameter vascular grafts currently used in cardiac and lower-extremity revascularization procedures. Adipose-derived, mesenchymal stem cell-infused, tissue-engineered vascular grafts have been shown to promote remodeling and vascular homeostasis in vivo and offer a possible treatment solution for those with cardiovascular disease. Unfortunately, the time needed to cultivate adipose-derived mesenchymal stem cells remains a large hurdle for tissue-engineered vascular grafts as a treatment option. The purpose of this study was to determine if stromal vascular fraction (known to contain progenitor cells) seeded tissue-engineered vascular grafts would remain patent in vivo and remodel, allowing for a "same-day" process for tissue-engineered vascular graft fabrication and implantation. METHODS Stromal vascular fraction, obtained from adult human adipose tissue, was seeded within 4 hours after acquisition from the patient onto poly(ester urethane)urea bilayered scaffolds using a customized rotational vacuum seeding device. Constructs were then surgically implanted as abdominal aortic interposition grafts in Lewis rats. RESULTS Findings revealed patency in 5 of 7 implanted scaffolds at 8 weeks, along with neotissue formation and remodeling occurring in patent tissue-engineered vascular grafts. Patency was documented using angiography and gross inspection, and remodeling and vascular components were detected using immunofluorescent chemistry. CONCLUSIONS A "same-day" cell-seeded, tissue-engineered vascular graft can remain patent after implantation in vivo, with neotissue formation and remodeling occurring by 8 weeks.
Collapse
Affiliation(s)
- Darren G Haskett
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Kamiel S Saleh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa
| | - Katherine L Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa
| | | | - Samuel K Luketich
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pa
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Pathology, University of Pittsburgh, Pittsburgh, Pa
| | - Lauren E Kokai
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Antonio D'Amore
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pa; RiMED Foundation, Palermo, Italy
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - William R Wagner
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pa; Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, Pa
| | - David A Vorp
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pa; Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, Pa; Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
28
|
A Novel Porcine Model for Future Studies of Cell-enriched Fat Grafting. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2018; 6:e1735. [PMID: 29876178 PMCID: PMC5977937 DOI: 10.1097/gox.0000000000001735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Cell-enriched fat grafting has shown promising results for improving graft survival, although many questions remain unanswered. A large animal model is crucial for bridging the gap between rodent studies and human trials. We present a step-by-step approach in using the Göttingen minipig as a model for future studies of cell-enriched large volume fat grafting. Methods: Fat grafting was performed as bolus injections and structural fat grafting. Graft retention was assessed by magnetic resonance imaging after 120 days. The stromal vascular fraction (SVF) was isolated from excised fat and liposuctioned fat from different anatomical sites and analyzed. Porcine adipose-derived stem/stromal cells (ASCs) were cultured in different growth supplements, and population doubling time, maximum cell yield, expression of surface markers, and differentiation potential were investigated. Results: Structural fat grafting in the breast and subcutaneous bolus grafting in the abdomen revealed average graft retention of 53.55% and 15.28%, respectively, which are similar to human reports. Liposuction yielded fewer SVF cells than fat excision, and abdominal fat had the most SVF cells/g fat with SVF yields similar to humans. Additionally, we demonstrated that porcine ASCs can be readily isolated and expanded in culture in allogeneic porcine platelet lysate and fetal bovine serum and that the use of 10% porcine platelet lysate or 20% fetal bovine serum resulted in population doubling time, maximum cell yield, surface marker profile, and trilineage differentiation that were comparable with humans. Conclusions: The Göttingen minipig is a feasible and cost-effective, large animal model for future translational studies of cell-enriched fat grafting.
Collapse
|
29
|
Griessl M, Buchberger AM, Regn S, Kreutzer K, Storck K. Uncultivated stromal vascular fraction is equivalent to adipose-derived stem and stromal cells on porous polyurethrane scaffolds forming adipose tissue in vivo. Laryngoscope 2018; 128:E206-E213. [PMID: 29446455 DOI: 10.1002/lary.27124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVES/HYPOTHESIS To find an alternative approach to contemporary techniques in tissue augmentation and reconstruction, tissue engineering strategies aim to involve adipose-derived stem and stromal cells (ASCs) harboring a strong differentiation potential into various tissue types such as bone, cartilage, and fat. STUDY DESIGN Animal research. METHODS The stromal vascular fraction (SVF) was used directly as a cell source to provide a potential alternative to contemporary ASC-based adipose tissue engineering. Seeded in TissuCol fibrin, we applied ASCs or SVF cells to porous, degradable polyurethane (PU) scaffolds. RESULTS We successfully demonstrated the in vivo generation of volume-stable, well-vascularized PU-based constructs containing host-derived mature fat pads. Seeded human stem cells served as modulators of host-cell migration rather than differentiating themselves. We further demonstrated that preliminary culture of SVF cells was not necessary. CONCLUSIONS Our results bring adipose tissue engineering, together with automated processing devices, closer to clinical applicability. The time-consuming and cost-intensive culture and induction of the ASCs is not necessary. LEVEL OF EVIDENCE NA. Laryngoscope, 128:E206-E213, 2018.
Collapse
Affiliation(s)
- Michael Griessl
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Anna-Maria Buchberger
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Sybille Regn
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Kilian Kreutzer
- Department of Maxillofacial Surgery, University Clinic of the Charité Berlin, Berlin, Germany
| | - Katharina Storck
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| |
Collapse
|