1
|
Arcani R, Velier M, Sabatier F, Simoncini S, Abellan-Lopez M, Granel B, Benyamine A, Gomes de Pinho Q, Dani V, Gentile P, Magalon G, Menkes S, Sampson S, Verpaele A, Vonk L, Magalon J, Daumas A. Nanofat Use in Regenerative Medicine: A Systematic Literature Review and Consensus Recommendations from Expert Opinions. Facial Plast Surg Aesthet Med 2025. [PMID: 40250993 DOI: 10.1089/fpsam.2024.0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2025] Open
Abstract
Objective: To report in vitro, preclinical, and clinical effectiveness of nanofat in adults undergoing reconstructive or functional surgery and to produce a series of consensus statements about nanofat definition by experts. Methods: We conducted a systematic review using PubMed and Web of Science database, retaining studies about nanofat alone. To produce consensus recommendations about nanofat, we invited experts to answer a survey about manufacturing, biological characteristics, and nomenclature of nanofat. Results: A review of 39 articles showed that nanofat seems to have strong regenerative potential. There were 16 studies about the clinical effectiveness of the nanofat in wound healing, aesthetic surgery, and functional disabilities. However, majority of applications lack robust clinical evidence, mainly due to the design of the clinical studies. The experts suggested that nanofat refers to lipoaspirate that benefits from a washing step, followed by emulsification (20-30 passes) with a connector size between 1.2 and 1.6 mm, and a final filtration step (pore size around 300-500 µm). Conclusion: Nanofat seems to have strong regenerative potentials but with a lack of robust clinical evidences. Our experts have suggested the first consensus about a definition of the nanofat that can be used by the academic societies in the coming years.
Collapse
Affiliation(s)
- Robin Arcani
- Internal Medicine and Therapeutics Department, CHU La Timone, Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
| | - Mélanie Velier
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC, Marseille, France
| | - Florence Sabatier
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC, Marseille, France
| | - Stéphanie Simoncini
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
| | - Maxime Abellan-Lopez
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Brigitte Granel
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord AP-HM, Marseille, France
| | - Audrey Benyamine
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord AP-HM, Marseille, France
| | - Quentin Gomes de Pinho
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord AP-HM, Marseille, France
| | | | - Pietro Gentile
- Department of Surgical Science, Medical School, "Tor Vergata" University, Rome, Italy
| | | | | | - Steve Sampson
- The Orthohealing Center, Los Angeles, California, USA
| | - Alexis Verpaele
- Tonnard and Verpaele Plastic Surgery Associates, Ghent, Belgium
| | - Lucienne Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands & Xintela AB, Lund, Sweden
| | - Jérémy Magalon
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC, Marseille, France
| | - Aurélie Daumas
- Internal Medicine and Therapeutics Department, CHU La Timone, Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France
- Center for Cardiovascular and Nutrition Research (C2VN), INRA 1260, INSERM UMR_S 1263, Aix-Marseille University, Marseille, France
| |
Collapse
|
2
|
Fu H, Wang C. Micro-fragmented adipose tissue-An innovative therapeutic approach: A narrative review. Medicine (Baltimore) 2025; 104:e41724. [PMID: 40020111 PMCID: PMC11875617 DOI: 10.1097/md.0000000000041724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Subcutaneous adipose tissue provides distinct advantages as a source of mesenchymal stem cells due to its accessibility and the ease of isolating stem cells. Human adipose stem cells, located in the stromal-vascular fraction, can be harvested using mechanical methods to produce microfragmented adipose tissue (MFAT). Local injections of MFAT have shown potential in promoting natural tissue regeneration. This review introduces the concept of MFAT, highlights its clinical applications, and explores its potential in regenerative medicine, offering insights into its role as an innovative therapeutic approach.
Collapse
Affiliation(s)
- Hongjuan Fu
- Department of Anesthesiology, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Congcong Wang
- Department of Joint Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
3
|
Richter DL, Harrison JL, Faber L, Schrader S, Zhu Y, Pierce C, Watson L, Shetty AK, Schenck RC. Microfragmented Adipose Tissue Injection Reduced Pain Compared With a Saline Control Among Patients With Symptomatic Osteoarthritis of the Knee During 1-Year Follow-Up: A Randomized Controlled Trial. Arthroscopy 2025; 41:248-260. [PMID: 39243998 DOI: 10.1016/j.arthro.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE To evaluate the effectiveness of microfragmented adipose tissue (MFAT) for pain relief and improved joint functionality in osteoarthritis (OA) of the knee in a randomized controlled clinical trial with 1-year follow-up. METHODS Seventy-five patients were stratified by baseline pain level and randomized to 1 of 3 treatment groups: MFAT, corticosteroid (CS), or saline control (C) injection. Patients 18 years of age or older, diagnosed with symptomatic OA of the knee, with radiographic evidence of OA of the knee and a visual analog pain scale score of 3 of 10 or greater were included. Patients were excluded if they had any previous intra-articular knee injection, current knee ligamentous instability, or an allergy to lidocaine/corticosteroid. The visual analog pain scale, Western Ontario and McMaster Universities Osteoarthritis Index, and the Knee Injury and Osteoarthritis Outcome score (KOOS) were recorded preprocedure and at 2 weeks, 6 weeks, 3 and 6 months, and 1-year follow-up. RESULTS MFAT demonstrated consistent and statistically significant improvements across all primary outcome measures for joint pain and functionality compared with C. For MFAT, there was a significant improvement over baseline at each follow-up, with median (95% confidence interval) KOOS Pain score changes of 18.1 (11.1-26.4) at week 2 to 27.8 (19.4-37.5) at 1 year. For CS, the median KOOS pain score reached a maximum of 22.2 (15.3-30.6) at week 2, only to level off to 13.9 (-2.8 to 29.2), a level not statistically different from baseline, at 1 year. The median changes for C hovered around 6 to 11 points, with statistically significant improvements over baseline indicating a placebo effect. Similar trends were seen for the Western Ontario and McMaster Universities Osteoarthritis Index Pain score and VAS Pain score. CONCLUSIONS In this study, MFAT demonstrated a clinically significant improvement in primary outcome scores compared with the C group, whereas the CS group only showed statistically significant improvement compared with the C group at 2 and 6 weeks. This finding indicates that MFAT may be a viable alternative treatment for patients with OA of the knee who fall into the orthopaedic treatment gap. LEVEL OF EVIDENCE Level II, partially blinded, randomized controlled clinical trial.
Collapse
Affiliation(s)
- Dustin L Richter
- Division of Sports Medicine, Department of Orthopaedics, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Joshua L Harrison
- Division of Plastic Surgery, Department of Surgery, University of New Mexico, Albuquerque, New Mexico, U.S.A..
| | - Lauren Faber
- Division of Urology, Department of Surgery, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Samuel Schrader
- Department of Orthopedics, Mayo Clinic, Rochester, Minnesota, U.S.A
| | - Yiliang Zhu
- Division of Epidemiology, Biostatistics, and Preventive Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Carina Pierce
- Division of Sports Medicine, Department of Orthopaedics, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Leorrie Watson
- Division of Sports Medicine, Department of Orthopaedics, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Anil K Shetty
- Division of Plastic Surgery, Department of Surgery, University of New Mexico, Albuquerque, New Mexico, U.S.A
| | - Robert C Schenck
- Division of Sports Medicine, Department of Orthopaedics, University of New Mexico, Albuquerque, New Mexico, U.S.A
| |
Collapse
|
4
|
McSweeney JE, Yong LY, Goddard NV, Wong JK. Does Secondary Mechanical Manipulation of Lipoaspirate Enhance the Vasculogenic Potential of Fat Grafts? A Systematic Review. Ann Plast Surg 2024; 93:389-396. [PMID: 39150855 DOI: 10.1097/sap.0000000000004048] [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: 08/18/2024]
Abstract
BACKGROUND Fat grafting is a highly versatile option in the reconstructive armamentarium but with unpredictable retention rates and outcomes. The primary outcome of this systematic review was to assess whether secondary mechanically processed lipoaspirate favorably enhances the vasculogenic potential of fat grafts when compared to unprocessed lipoaspirate or fat grafts prepared using centrifugation alone. The secondary outcome was to assess the evidence around graft retention and improved outcomes when comparing the aforementioned groups. METHODS A search on MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials was conducted up to February 2022. All human and animal research, which provided a cross-comparison between unprocessed, centrifuged, secondary mechanically fragmented (SMF) or secondary mechanically disrupted (SMD) fat grafts, was included. RESULTS Thirty-one full texts were included. Vasculogenic potential was assessed by quantification of angiogenic growth factors and cellular composition. Cellular composition of mesenchymal stem cells, perivascular stem cells, and endothelial progenitor cells was quantified by fluorescence activated cell sorting (FACS) analysis. Fat graft volume retention rates and fat grafting to aid wound healing were assessed. Although the presence of industry-funded studies and inadequate reporting of methodological data in some studies were sources of bias, data showed SMF grafts contain an enriched pericyte population with increased vascular endothelial growth factor (VEGF) secretion. Animal studies indicate that SMD grafts may increase rates of fat graft retention and wound closure compared to centrifuged grafts; however, clinical studies are yet to show similar results. CONCLUSIONS In this systematic review, we were able to conclude that the existing literature suggests mechanically processing fat, whether it be through fragmentation or disruption, improves vasculogenic potential by enhancing angiogenic growth factor and relevant vascular progenitor cell levels. Whilst in vivo animal studies are scarce, the review findings suggest that secondary mechanically processed fat enhances fat graft retention and can aid with wound healing. Further clinical studies are required to assess potential differences in human studies.
Collapse
Affiliation(s)
- Jared Ethan McSweeney
- From the Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, M13 9PL, UK
| | | | | | | |
Collapse
|
5
|
Guillaumes S, Hidalgo NJ, Bachero I, Pena R, Nogueira ST, Ardid J, Pera M. Efficacy of injection of autologous adipose tissue in the treatment of patients with complex and recurrent fistula-in-ano of cryptoglandular origin. Tech Coloproctol 2024; 28:81. [PMID: 38980511 PMCID: PMC11233338 DOI: 10.1007/s10151-024-02963-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/08/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Adipose tissue injections, a rich source of mesenchymal stem cells, have been successfully used to promote anal fistula healing. This study aimed to investigate the efficacy of adipose tissue injection in treating patients with complex and recurrent fistulas of cryptoglandular origin. METHODS We conducted a prospective, single-center, open-label, non-randomized, interventional clinical trial from January 2020 to December 2022. We enrolled nine patients, who were evaluated after at least 12 months of follow-up. All patients had seton removal, fistula tract excision or curettage, and a mucosal flap if possible or, alternatively, an internal opening suture. We used a commercially available system to collect and process adipose tissue prior to injection. This system allowed the collection, microfragmentation, and filtration of tissue. RESULTS Selected cases included six men and three women with a median age of 42 (range 31-55) years. All patients had an extended disease course period, ranging from 3 to 13 (mean 6.6) years, and a history of multiple previous surgeries, including two to eight interventions (a mean of 4.4 per case). All fistulas were high transsphincteric, four cases horseshoe and two cases with secondary suprasphincteric or peri-elevator tract fistulas. Six cases (66%) achieved complete fistula healing at a mean follow-up of 18 (range 12-36) months. Three cases (33.3%) experienced reduced secretion and decreased anal discomfort. CONCLUSIONS In patients with complex and recurrent fistulas, such as the ones described, many from palliative treatments with setons, the adjuvant injection of adipose tissue might help achieve complete healing or improvement in a significant percentage of cases. CLINICALTRIALS The study protocol was prospectively registered on ClinicalTrials.gov (NCT04750499).
Collapse
Affiliation(s)
| | - N J Hidalgo
- Hospital Clinic de Barcelona, Barcelona, Spain.
| | - I Bachero
- Hospital Clinic de Barcelona, Barcelona, Spain
| | - R Pena
- Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - J Ardid
- Hospital Clinic de Barcelona, Barcelona, Spain
| | - M Pera
- Hospital Clinic de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Malisetyan T, Harrison JL, Shahriari SR, Clarke TN, Rogol EV, Borah GL. Autologous Fat Transfer in Craniofacial Surgery. FACE 2024; 5:279-291. [DOI: 10.1177/27325016241238441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Over the past two decades, autologous fat transfer has garnered significant recognition and widespread adoption within esthetic and reconstructive surgical domains. In craniofacial surgery, fat transplantation is frequently employed to address soft tissue volumetric deficiencies and asymmetries that influence facial contours. While adipose tissue (AT) is widely regarded as an optimal choice for augmentation due to its abundant availability and biocompatibility, the unpredictability and heightened resorption rates observed with traditional lipofilling techniques present a challenge for clinicians. Adipose-derived stem cells (ASCs) housed within the grafted tissue play a pivotal role in graft survival and offer avenues for tissue repair due to their angiogenic, anti-inflammatory, and immunosuppressive properties. Micro Fragmentation of Adipose Tissue (MFAT), utilized in several FDA-approved processing devices, has demonstrated promising outcomes in treating osteoarthritic joints, with success primarily attributed to enhanced paracrine function of ASCs via preservation of the perivascular niche. Currently, its application for treating bone or articular defects in the craniofacial region, including abnormalities of the temporomandibular joint, remains limited. This scarcity underscores the need for further investigation prior to its widespread integration into clinical practice.
Collapse
Affiliation(s)
- Tatevik Malisetyan
- Nova Southeastern University, Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, FL, USA
| | | | | | - Tegan N. Clarke
- University of New Mexico School of Medicine, Albuquerque, NM, USA
| | | | - Gregory L. Borah
- University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
7
|
Epanomeritakis IE, Khan WS. Adipose-derived regenerative therapies for the treatment of knee osteoarthritis. World J Stem Cells 2024; 16:324-333. [PMID: 38690511 PMCID: PMC11056639 DOI: 10.4252/wjsc.v16.i4.324] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 04/25/2024] Open
Abstract
Knee osteoarthritis is a degenerative condition with a significant disease burden and no disease-modifying therapy. Definitive treatment ultimately requires joint replacement. Therapies capable of regenerating cartilage could significantly reduce financial and clinical costs. The regenerative potential of mesenchymal stromal cells (MSCs) has been extensively studied in the context of knee osteoarthritis. This has yielded promising results in human studies, and is likely a product of immunomodulatory and chondroprotective biomolecules produced by MSCs in response to inflammation. Adipose-derived MSCs (ASCs) are becoming increasingly popular owing to their relative ease of isolation and high proliferative capacity. Stromal vascular fraction (SVF) and micro-fragmented adipose tissue (MFAT) are produced by the enzymatic and mechanical disruption of adipose tissue, respectively. This avoids expansion of isolated ASCs ex vivo and their composition of heterogeneous cell populations, including immune cells, may potentiate the reparative function of ASCs. In this editorial, we comment on a multicenter randomized trial regarding the efficacy of MFAT in treating knee osteoarthritis. We discuss the study's findings in the context of emerging evidence regarding adipose-derived regenerative therapies. An underlying mechanism of action of ASCs is proposed while drawing important distinctions between the properties of isolated ASCs, SVF, and MFAT.
Collapse
Affiliation(s)
- Ilias E Epanomeritakis
- Division of Trauma and Orthopaedic Surgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Wasim S Khan
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
8
|
Bonnet M, Ertlen C, Seblani M, Brezun JM, Coyle T, Cereda C, Zuccotti G, Colli M, Desouches C, Decherchi P, Carelli S, Marqueste T. Activated Human Adipose Tissue Transplantation Promotes Sensorimotor Recovery after Acute Spinal Cord Contusion in Rats. Cells 2024; 13:182. [PMID: 38247873 PMCID: PMC10814727 DOI: 10.3390/cells13020182] [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: 11/23/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) often result in sensory, motor, and vegetative function loss below the injury site. Although preclinical results have been promising, significant solutions for SCI patients have not been achieved through translating repair strategies to clinical trials. In this study, we investigated the effective potential of mechanically activated lipoaspirated adipose tissue when transplanted into the epicenter of a thoracic spinal contusion. Male Sprague Dawley rats were divided into three experimental groups: SHAM (uninjured and untreated), NaCl (spinal cord contusion with NaCl application), and AF (spinal cord contusion with transplanted activated human fat). Pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) were measured to assess endogenous inflammation levels 14 days after injury. Sensorimotor recovery was monitored weekly for 12 weeks, and gait and electrophysiological analyses were performed at the end of this observational period. The results indicated that AF reduced endogenous inflammation post-SCI and there was a significant improvement in sensorimotor recovery. Moreover, activated adipose tissue also reinstated the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions. This investigation highlights the efficacy of activated adipose tissue grafting in acute SCI, suggesting it is a promising therapeutic approach for spinal cord repair after traumatic contusion in humans.
Collapse
Affiliation(s)
- Maxime Bonnet
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Céline Ertlen
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Mostafa Seblani
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Jean-Michel Brezun
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Thelma Coyle
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
- Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Mattia Colli
- Podgora7 Clinic, Via Podgora 7, 20122 Milano, Italy
| | - Christophe Desouches
- Clinique Phénicia—CD Esthétique, 5 Boulevard Notre Dame, F-13006 Marseille, France
| | - Patrick Decherchi
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
| | - Tanguy Marqueste
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| |
Collapse
|
9
|
Sciarretta FV, Ascani C, Sodano L, Fossati C, Campisi S. One-stage cartilage repair using the autologous matrix-induced chondrogenesis combined with simultaneous use of autologous adipose tissue graft and adipose tissue mesenchymal cells technique: clinical results and magnetic resonance imaging evaluation at five-year follow-up. INTERNATIONAL ORTHOPAEDICS 2024; 48:267-277. [PMID: 37656198 PMCID: PMC10766726 DOI: 10.1007/s00264-023-05921-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/30/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE To evaluate medium-term outcomes of knee cartilage defects repair by autologous matrix-induced chondrogenesis combined with simultaneous use of autologous adipose tissue graft and adipose tissue mesenchymal cells, defined as LIPO-AMIC technique. METHODS The LIPO-AMIC technique has been used in ICRS degree III-IV knee defects. Eighteen patients have been prospectively evaluated during two and five years both clinically and by MRI. RESULTS Patients showed progressive significant improvement of all scores starting early at six months, and further increased values were noted till the last follow-up at 60 months. Mean subjective pre-operative IKDC score of 36.1 significantly increased to 86.4 at 24 months and to 87.2 at 60 months. Mean pre-operative Lysholm score of 44.4 reached 93.5 at two years and 93.5 at five years. MRI examination showed early subchondral lamina regrowth and progressive maturation of repair tissue and filling of defects. The mean total MOCART score showed that a significative improvement from two year follow-up (69.1 points) to last follow-up was 81.9 points (range, 30-100 points, SD 24). Complete filling of the defect at the level of the surrounding cartilage was found in 77.8%. CONCLUSIONS Adipose tissue can represent ideal source of MSCs since easiness of withdrawal and definite chondrogenic capacity. This study clearly demonstrated the LIPO-AMIC technique to be feasible for treatment of knee cartilage defects and to result in statistically significant progressive clinical, functional and pain improvement in all treated patients better than what reported for the AMIC standard technique, starting very early from the 6-month follow-up and maintaining the good clinical results more durably with stable results at mid-term follow-up.
Collapse
Affiliation(s)
- Fabio Valerio Sciarretta
- Clinica Nostra Signora della Mercede, Via Tagliamento 25, 00198, Rome, Italy.
- Accademia Biomedica Rigenerativa (ABRI), Via Misurina 56, 00135, Rome, Italy.
- Artemisia Lab, Via Piave 76, 00198, Rome, Italy.
| | | | - Luca Sodano
- Ospedale San Luca, Via Francesco Cammarota, 84078, Vallo della Lucania, SA, Italy
| | - Carolina Fossati
- Accademia Biomedica Rigenerativa (ABRI), Via Misurina 56, 00135, Rome, Italy
- Artemisia Lab, Via Piave 76, 00198, Rome, Italy
| | - Silvana Campisi
- Accademia Biomedica Rigenerativa (ABRI), Via Misurina 56, 00135, Rome, Italy
- Artemisia Lab, Via Piave 76, 00198, Rome, Italy
| |
Collapse
|
10
|
Wu CZ, Shi ZY, Wu Z, Lin WJ, Chen WB, Jia XW, Xiang SC, Xu HH, Ge QW, Zou KA, Wang X, Chen JL, Wang PE, Yuan WH, Jin HT, Tong PJ. Mid-term outcomes of microfragmented adipose tissue plus arthroscopic surgery for knee osteoarthritis: A randomized, active-control, multicenter clinical trial. World J Stem Cells 2023; 15:1063-1076. [PMID: 38179213 PMCID: PMC10762526 DOI: 10.4252/wjsc.v15.i12.1063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent form of degenerative whole-joint disease. Before the final option of knee replacement, arthroscopic surgery was the most widely used joint-preserving surgical treatment. Emerging regenerative therapies, such as those involving platelet-rich plasma, mesenchymal stem cells, and microfragmented adipose tissue (MFAT), have been pushed to the forefront of treatment to prevent the progression of OA. Currently, MFAT has been successfully applied to treat different types of orthopedic diseases. AIM To assess the efficacy and safety of MFAT with arthroscopic surgery in patients with knee OA (KOA). METHODS A randomized, multicenter study was conducted between June 2017 and November 2022 in 10 hospitals in Zhejiang, China. Overall, 302 patients diagnosed with KOA (Kellgren-Lawrence grades 2-3) were randomized to the MFAT group (n = 151, were administered MFAT following arthroscopic surgery), or the control group (n = 151, were administered hyaluronic acid following arthroscopic surgery). The study outcomes were changes in the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, the visual analog scale (VAS) score, the Lequesne index score, the Whole-Organ Magnetic Resonance Imaging Score (WORMS), and safety over a 24-mo period from baseline. RESULTS The changes in the WOMAC score (including the three subscale scores), VAS pain score, and Lequesne index score at the 24-mo mark were significantly different in the MFAT and control groups, as well as when comparing values at the posttreatment visit and those at baseline (P < 0.001). The MFAT group consistently demonstrated significant decreases in the WOMAC pain scores and VAS scores at all follow-ups compared to the control group (P < 0.05). Furthermore, the WOMAC stiffness score, WOMAC function score, and Lequesne index score differed significantly between the groups at 12 and 24 mo (P < 0.05). However, no significant between-group differences were observed in the WORMS at 24 mo (P = 0.367). No serious adverse events occurred in both groups. CONCLUSION The MFAT injection combined with arthroscopic surgery treatment group showed better mid-term clinical outcomes compared to the control group, suggesting its efficacy as a therapeutic approach for patients with KOA.
Collapse
Affiliation(s)
- Cong-Zi Wu
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zhen-Yu Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Zhen Wu
- Department of Orthopaedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Wen-Jun Lin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang Province, China
| | - Wei-Bo Chen
- Department of Orthopaedic Surgery, Ruian Hospital of Traditional Chinese Medicine, Wenzhou 325299, Zhejiang Province, China
| | - Xue-Wen Jia
- Department of Orthopaedics, Ningbo First Hospital, Ningbo 315010, Zhejiang Province, China
| | - Si-Cheng Xiang
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Hui-Hui Xu
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Qin-Wen Ge
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai-Ao Zou
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Xu Wang
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jia-Li Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Ping-Er Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Wen-Hua Yuan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Hong-Ting Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Pei-Jian Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China.
| |
Collapse
|
11
|
Veronesi F, Andriolo L, Salerno M, Boffa A, Giavaresi G, Filardo G. Adipose Tissue-Derived Minimally Manipulated Products versus Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: A Systematic Review of Clinical Evidence and Meta-Analysis. J Clin Med 2023; 13:67. [PMID: 38202074 PMCID: PMC10780289 DOI: 10.3390/jcm13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
The use of minimally manipulated adipose tissue (MM-AT) products is gaining increasing interest for the treatment of knee osteoarthritis (OA). MM-AT represents an easy way to exploit adipose tissue properties, although clinical evidence is still limited, as well as their benefits with respect to more documented orthobiologics like platelet-rich plasma (PRP). A systematic review and meta-analysis were performed to evaluate the safety and efficacy of MM-AT products for knee OA management. The risk of bias of the included studies was evaluated using the Dawns and Black checklist for all the included studies and RoB-2.0 for randomized controlled trials (RCTs). Thirty-three clinical studies were included in the qualitative analysis: 13 prospective case series, 10 retrospective case series, 7 RCTs, 2 retrospective comparative studies, and 1 prospective comparative study. An overall clinical improvement and few minor adverse events were observed. Five RCTs comparing MM-AT and PRP injections were meta-analyzed, showing comparable results. The analysis also highlighted the limits of the literature, with only a few high-level trials and an overall low quality. Even though the current literature is still limited, the available evidence suggests the safety and overall positive results of the intra-articular injections of MM-AT products for knee OA treatment.
Collapse
Affiliation(s)
- Francesca Veronesi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.V.); (G.G.)
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Manuela Salerno
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.S.)
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.V.); (G.G.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.S.)
| |
Collapse
|
12
|
Cavallo C, Boffa A, Salerno M, Merli G, Grigolo B, Filardo G. Adipose Tissue-Derived Products May Present Inflammatory Properties That Affect Chondrocytes and Synoviocytes from Patients with Knee Osteoarthritis. Int J Mol Sci 2023; 24:12401. [PMID: 37569775 PMCID: PMC10418602 DOI: 10.3390/ijms241512401] [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: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Adipose tissue-derived cell-based injectable therapies have been demonstrated to have disease-modifying effects on joint tissues in preclinical studies on animal osteoarthritis (OA) models, but clinical results are heterogeneous and not always satisfactory. The aim of this study was to investigate the influence of adipose tissue properties on the therapeutic effects of the adipose-derived product in an in vitro OA setting. Micro-fragmented adipose tissue (MF-AT) samples were obtained from 21 OA patients (mean age 51.7 ± 11.8 years, mean BMI 25.7 ± 4.1 kg/m2). The analysis of the MF-AT supernatant was performed to analyze the release of inflammatory factors. The effects of MF-AT inflammatory factors were investigated on chondrocytes and synoviocytes gene expression levels. Patients' characteristics were analyzed to explore their influence on MF-AT inflammatory molecules and on the MF-AT effects on the gene expression of chondrocytes and synoviocytes. The study results demonstrated that adipose tissue-derived products may present inflammatory properties that influence the therapeutic potential for OA treatment, with products with a higher pro-inflammatory profile stimulating a higher expression of genes related to a more inflamed and catabolic phenotype. A higher pro-inflammatory cytokine pattern and a higher pro-inflammatory effect were found in adipose tissue-derived products obtained from OA patients with higher BMI.
Collapse
Affiliation(s)
- Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (C.C.); (B.G.)
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Manuela Salerno
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| | - Giulia Merli
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (C.C.); (B.G.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| |
Collapse
|
13
|
Petrova V, Vachkova E. Outlook of Adipose-Derived Stem Cells: Challenges to Their Clinical Application in Horses. Vet Sci 2023; 10:vetsci10050348. [PMID: 37235430 DOI: 10.3390/vetsci10050348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Adipose tissue is recognized as the major endocrine organ, potentially acting as a source of mesenchymal stem cells for various applications in regenerative medicine. Athletic horses are often exposed to traumatic injuries, resulting in severe financial losses. The development of adipose-derived stem cells' regenerative potential depends on many factors. The extraction of stem cells from subcutaneous adipose tissue is non-invasive, non-traumatic, cheaper, and safer than other sources. Since there is a lack of unique standards for identification, the isolated cells and applied differentiation protocols are often not species-specific; therefore, the cells cannot reveal their multipotent properties, so their stemness features remain questionable. The current review discusses some aspects of the specificity of equine adipose stem cells concerning their features, immunophenotyping, secretome profile, differentiation abilities, culturing conditions, and consequent possibilities for clinical application in concrete disorders. The presented new approaches elucidate the possibility of the transition from cell-based to cell-free therapy with regenerative purposes in horses as an alternative treatment to cellular therapy. In conclusion, their clinical benefits should not be underestimated due to the higher yield and the physiological properties of adipose-derived stem cells that facilitate the healing and tissue regeneration process and the ability to amplify the effects of traditional treatments. More profound studies are necessary to apply these innovative approaches when treating traumatic disorders in racing horses.
Collapse
Affiliation(s)
- Valeria Petrova
- Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Ekaterina Vachkova
- Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| |
Collapse
|
14
|
Scattini G, Pellegrini M, Severi G, Cagiola M, Pascucci L. The Stromal Vascular Fraction from Canine Adipose Tissue Contains Mesenchymal Stromal Cell Subpopulations That Show Time-Dependent Adhesion to Cell Culture Plastic Vessels. Animals (Basel) 2023; 13:ani13071175. [PMID: 37048431 PMCID: PMC10093060 DOI: 10.3390/ani13071175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (MSCs) are extensively studied in both human and veterinary medicine. Their isolation is usually performed by collagenase digestion followed by filtration and removal of nonadherent tissue remnants 48 h after seeding. We observed that waste tissue fragments contain cells that adhere belatedly to the plastic. We aimed to investigate their basic properties to speculate on the possible existence of MSC subpopulations. Adipose tissue from three dogs was enzymatically digested. Three cell populations that adhered to the culture plastic 48, 96, and 144 h after seeding were obtained. After expansion, they were analyzed by flow cytometry for MSC-positive (CD90, CD44, and CD29) and -negative (CD14, MHCII, and CD45) markers as well as for endothelial, pericyte, and smooth muscle cell markers (CD31, CD146, and alpha-SMA). Furthermore, cells were assessed for viability, doubling time, and trilineage differentiation ability. No significant differences were found between the three subpopulations. As a result, this procedure has proven to be a valuable method for dramatically improving MSCs yield. As a consequence of cell recovery optimization, the amount of tissue harvested could be reduced, and the time required to obtain sufficient cells for clinical applications could be shortened. Further studies are needed to uncover possible different functional properties.
Collapse
Affiliation(s)
- Gabriele Scattini
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
| | - Martina Pellegrini
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy
- Correspondence: (M.P.); (L.P.); Tel.: +39-075-3431 (M.P.); +39-075-585-7632 (L.P.)
| | - Giulio Severi
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy
| | - Monica Cagiola
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
- Correspondence: (M.P.); (L.P.); Tel.: +39-075-3431 (M.P.); +39-075-585-7632 (L.P.)
| |
Collapse
|
15
|
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: 22] [Impact Index Per Article: 7.3] [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
|
16
|
Zaffagnini S, Andriolo L, Boffa A, Poggi A, Cenacchi A, Busacca M, Kon E, Filardo G, Di Martino A. Microfragmented Adipose Tissue Versus Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: A Prospective Randomized Controlled Trial at 2-Year Follow-up. Am J Sports Med 2022; 50:2881-2892. [PMID: 35984721 DOI: 10.1177/03635465221115821] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Intra-articular microfragmented adipose tissue (MF-AT) injections have been proposed for the treatment of knee osteoarthritis (OA). PURPOSE To compare a single injection of MF-AT or platelet-rich plasma (PRP) in terms of clinical outcomes and OA progression. STUDY DESIGN Randomized controlled trial; Level of evidence, 1. METHODS A total of 118 patients with symptomatic knee OA were randomized to receive a single intra-articular injection of MF-AT or PRP. Patients were evaluated before the injection and at 1, 3, 6, 12, and 24 months with the International Knee Documentation Committee (IKDC) subjective score, Knee injury and Osteoarthritis Outcome Score (KOOS) subscales, EuroQol visual analogue scale (EQ-VAS), EuroQol 5 dimensions (EQ-5D), and visual analogue scale (VAS) for pain. Primary outcomes were the IKDC subjective score and the KOOS pain subscore at 6 months. Knees were evaluated at baseline and at 6, 12, and 24 months with radiography and high-resolution magnetic resonance imaging (MRI) using the Whole-Organ Magnetic Resonance Imaging Score (WORMS). RESULTS Both MF-AT and PRP provided a statistically and clinically significant improvement up to 24 months. The improvement in the IKDC subjective score from baseline to 6 months was similar in both MF-AT (41.1 ± 16.3 to 57.3 ± 18.8) and PRP (44.8 ± 17.3 to 58.4 ± 18.1) groups (P < .0005). The improvement in the KOOS pain subscore from baseline to 6 months was similar in both the MF-AT (58.4 ± 15.9 to 75.8 ± 17.4) and PRP (63.5 ± 17.8 to 75.5 ± 16.1) groups (P < .0005). Overall, no differences were found between the MF-AT and PRP groups in terms of clinical outcomes, adverse events (18.9% and 10.9%, respectively), and failures (15.1% and 25.5%, respectively). Radiographic and MRI findings did not show changes after the injection. As a secondary outcome, more patients in the MF-AT group with moderate/severe OA reached the minimal clinically important difference for the IKDC score at 6 months compared with the PRP group (75.0% vs 34.6%, respectively; P = .005). CONCLUSION A single intra-articular injection of MF-AT was not superior to PRP, with comparable low numbers of failures and adverse events and without disease progression. No differences were found in clinical and imaging results between the 2 biological approaches.
Collapse
Affiliation(s)
- Stefano Zaffagnini
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alberto Poggi
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Annarita Cenacchi
- Servizio Trasfusionale Unico Metropolitano, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Elizaveta Kon
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Italy
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandro Di Martino
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
17
|
Zhang S, Li J, Li C, He J, Ling F, Liu G. Isolation and identification of a mesenchymal stem/stromal cell-like population from pediatric urethral tissue. In Vitro Cell Dev Biol Anim 2022; 58:503-511. [PMID: 35817989 DOI: 10.1007/s11626-022-00697-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/23/2022] [Indexed: 11/25/2022]
Abstract
Mesenchymal stem cells (MSCs) are important seed cells for cell therapy and tissue engineering because of their multidirectional differentiation potential, high proliferative capacity, low immunogenicity, and immunomodulatory ability. In this study, we successfully isolated and cultured a population of mesenchymal stem-like cells from pediatric urethra (PU-MSLCs). The cells had a spindle-shaped fibroblast-like morphology, similar to MSCs derived from other tissues. The PU-MSLCs highly expressed MSC surface markers CD29, CD73, CD90, and CD105 but were negative for leukocyte common antigen CD45, and MHC class II-encoded molecule HLA-DR. After in vitro induction, the PU-MSLCs had the potential to differentiate into adipocytes, osteocytes, and chondrocytes. The PU-MSLCs maintained a normal karyotype and showed no tumorigenicity during long-term cultivation. We thus demonstrated that the mesenchymal stem/stromal cell-like population obtained from pediatric urethra tissue is capable of self-renewal and multidirectional differentiation, has promising application prospects for cell therapy and tissue engineering, and is expected to contribute to urethral tissue reconstruction.
Collapse
Affiliation(s)
- Shilin Zhang
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China.
| | - Jierong Li
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Chunjing Li
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Jun He
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Fengsheng Ling
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Guoqing Liu
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| |
Collapse
|
18
|
Screpis D, Natali S, Farinelli L, Piovan G, Iacono V, de Girolamo L, Viganò M, Zorzi C. Autologous Microfragmented Adipose Tissue for the Treatment of Knee Osteoarthritis: Real-World Data at Two Years Follow-Up. J Clin Med 2022; 11:jcm11051268. [PMID: 35268359 PMCID: PMC8911134 DOI: 10.3390/jcm11051268] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 12/10/2022] Open
Abstract
The purpose of the present study was to assess, prospectively, the safety, clinical effectiveness, and feasibility of a single intra-articular injection of microfragmented adipose tissue in different stages of knee osteoarthritis (OA). The study included patients (aged 18−70 years), affected by OA (Kellgren−Lawrence I-IV). Unselected patients were evaluated before and prospectively after 6, 12, and 24 months from the injection. Visual analog scale (VAS) and knee injury and osteoarthritis outcome score (KOOS) were used for clinical evaluations. A total of 202 patients were eligible. The mean follow-up time in the cohort of patients was 24.5 ± 9.6 months. Total KOOS significantly improved from pre-operative baseline levels to 6-month follow-up (p < 0.001), and again between 6- and 12-month follow-ups (p < 0.001). The VAS showed a prompt reduction at 6 months (p < 0.001 vs. baseline), but then it increased again at 12 months compared to the 6-month assessment (p < 0.001), even though it remained lower than baseline (p < 0.001). At 24 months, patients with KL-IV demonstrated a lower improvement compared to baseline; patients that had undergone previous corticosteroid injections had a greater risk to further injection treatment. The collected clinical results suggest that MFAT may represent a safe and effective treatment for OA symptoms, offering a low-demanding and minimally invasive treatment.
Collapse
Affiliation(s)
- Daniele Screpis
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (D.S.); (G.P.); (V.I.); (C.Z.)
| | - Simone Natali
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (D.S.); (G.P.); (V.I.); (C.Z.)
- Correspondence:
| | - Luca Farinelli
- Clinical Ortopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60020 Ancona, Italy;
| | - Gianluca Piovan
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (D.S.); (G.P.); (V.I.); (C.Z.)
| | - Venanzio Iacono
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (D.S.); (G.P.); (V.I.); (C.Z.)
| | - Laura de Girolamo
- Orthopaedic Biotechnology Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (L.d.G.); (M.V.)
| | - Marco Viganò
- Orthopaedic Biotechnology Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (L.d.G.); (M.V.)
| | - Claudio Zorzi
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (D.S.); (G.P.); (V.I.); (C.Z.)
| |
Collapse
|
19
|
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]
|
20
|
Ferroni L, De Francesco F, Pinton P, Gardin C, Zavan B. Methods to isolate adipose tissue-derived stem cells. Methods Cell Biol 2022; 171:215-228. [DOI: 10.1016/bs.mcb.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Stem Cells in Autologous Microfragmented Adipose Tissue: Current Perspectives in Osteoarthritis Disease. Int J Mol Sci 2021; 22:ijms221910197. [PMID: 34638538 PMCID: PMC8508703 DOI: 10.3390/ijms221910197] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a chronic debilitating disorder causing pain and gradual degeneration of weight-bearing joints with detrimental effects on cartilage volume as well as cartilage damage, generating inflammation in the joint structure. The etiology of OA is multifactorial. Currently, therapies are mainly addressing the physical and occupational aspects of osteoarthritis using pharmacologic pain treatment and/or surgery to manage the symptomatology of the disease with no specific regard to disease progression or prevention. Herein, we highlight alternative therapeutics for OA specifically considering innovative and encouraging translational methods with the use of adipose mesenchymal stem cells.
Collapse
|
22
|
Rey F, Messa L, Pandini C, Barzaghini B, Micheletto G, Raimondi MT, Bertoli S, Cereda C, Zuccotti GV, Cancello R, Carelli S. Transcriptional characterization of subcutaneous adipose tissue in obesity affected women highlights metabolic dysfunction and implications for lncRNAs. Genomics 2021; 113:3919-3934. [PMID: 34555498 DOI: 10.1016/j.ygeno.2021.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
Obesity is a complex disease with multifactorial causes, and its prevalence is becoming a serious health crisis. For this reason, there is a crucial need to identify novel targets and players. With this aim in mind, we analyzed via RNA-sequencing the subcutaneous adipose tissue of normal weight and obesity-affected women, highlighting the differential expression in the two tissues. We specifically focused on long non-coding RNAs, as 6 of these emerged as dysregulated in the diseased-tissue (COL4A2-AS2, RPS21-AS, PELATON, ITGB2-AS1, ACER2-AS and CTEPHA1). For each of them, we performed both a thorough in silico dissection and in vitro validation, to predict their function during adipogenesis. We report the lncRNAs expression during adipose derived stem cells differentiation to adipocytes as model of adipogenesis and their potential modulation by adipogenesis-related transcription factors (C/EBPs and PPARγ). Moreover, inhibiting CTEPHA1 expression we investigated its impact on adipogenesis-related transcription factors, showing its significative dysregulation of C/EBPα expression. Lastly, we dissected the subcellular localization, pathway involvement and disease-correlation for coding differentially expressed genes. Together, these findings highlight a transcriptional deregulation at the basis of obesity, impacted by both coding and long non-coding RNAs.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy
| | - Letizia Messa
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Cecilia Pandini
- Genomic and post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Giancarlo Micheletto
- Department of Pathophysiology and Transplantation, INCO, Department of General Surgery, Istituto Clinico Sant'Ambrogio, University of Milan, Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Simona Bertoli
- Obesity Unit, Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy; International Center for the Assessment of Nutritional Status (ICANS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy; Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Raffaella Cancello
- Obesity Unit, Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy.
| |
Collapse
|
23
|
Muñoz MF, Argüelles S, Marotta F, Barbagallo M, Cano M, Ayala A. Effect of Age and Lipoperoxidation in Rat and Human Adipose Tissue-Derived Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6473279. [PMID: 33425211 PMCID: PMC7775166 DOI: 10.1155/2020/6473279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
A wide range of clinical applications in regenerative medicine were opened decades ago with the discovery of adult stem cells. Highly promising adult stem cells are mesenchymal stem/stromal cells derived from adipose tissue (ADSCs), primarily because of their abundance and accessibility. These cells have multipotent properties and have been used extensively to carry out autologous transplants. However, the biology of these cells is not entirely understood. Among other factors, the regeneration capacity of these cells will depend on both their capacity of proliferation/differentiation and the robustness of the biochemical pathways that allow them to survive under adverse conditions like those found in damaged tissues. The transcription factors, such as Nanog and Sox2, have been described as playing an important role in stem cell proliferation and differentiation. Also, the so-called longevity pathways, in which AMPK and SIRT1 proteins play a crucial role, are essential for cell homeostasis under stressful situations. These pathways act by inhibiting the translation through downregulation of elongation factor-2 (eEF2). In order to deepen knowledge of mesenchymal stem cell biology and which factors are determinant in the final therapeutic output, we evaluate in the present study the levels of all of these proteins in the ADSCs from humans and rats and how these levels are affected by aging and the oxidative environment. Due to the effect of aging and oxidative stress, our results suggest that before performing a cell therapy with ADSCs, several aspects reported in this study such as oxidative stress status and proliferation and differentiation capacity should be assessed on these cells. This would allow us to know the robustness of the transplanted cells and to predict the therapeutic result, especially in elder patients, where probably ADSCs do not carry out their biological functions in an optimal way.
Collapse
Affiliation(s)
- Mario F. Muñoz
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Sandro Argüelles
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Francesco Marotta
- ReGenera R&D International for Aging Intervention & Vitality Therapeutics, San Babila Clinic, Milan, Italy
| | - Mario Barbagallo
- Department of Geriatrics and Internal Medicine, University of Palermo, Italy
| | - Mercedes Cano
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Antonio Ayala
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| |
Collapse
|
24
|
Vinet-Jones H, F Darr K. Clinical use of autologous micro-fragmented fat progressively restores pain and function in shoulder osteoarthritis. Regen Med 2020; 15:2153-2161. [PMID: 33275470 DOI: 10.2217/rme-2020-0069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: We aim to show that the use of nondigested micro-fragmented adipose tissue (MFat™, Lipogems®) is a viable alternative for treatment of joint pain and inflammation associated dysfunction in shoulder osteoarthritis (OA). Materials & methods: A total of 25 subjects with OA received an injection of MFat™ and were followed at 6, 18 and 52 weeks intervals. Quantitative analysis of pain and function modalities were performed using the visual analog scale and the disabilities of the arm, shoulder and hand, respectively. Results: All study participants reported significant progressive improvement (p < 0.001) from baseline in visual analog scale and disabilities of the arm, shoulder and hand in shoulder OA cases up to a year post. Conclusion: MFat™ therapy improves pain and function in patients with shoulder OA and can provide a long-term alternative to surgical intervention.
Collapse
Affiliation(s)
- Heather Vinet-Jones
- Regenerative Medicine Covington, Orthopedic Sports Medicine Institute, Covington, LA 70433, USA
| | - Kevin F Darr
- Regenerative Medicine Covington, Orthopedic Sports Medicine Institute, Covington, LA 70433, USA
| |
Collapse
|
25
|
Frank RM, Bradsell H, Dragoo J. Adipose Derived Cellular Therapies–Arthroscopic Approaches. OPER TECHN SPORT MED 2020. [DOI: 10.1016/j.otsm.2020.150779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Al Haj G, Rey F, Giallongo T, Colli M, Marzani B, Giuliani G, Gorio A, Zuccotti GV, Di Giulio AM, Carelli S. A New Selective PPARγ Modulator Inhibits Triglycerides Accumulation during Murine Adipocytes' and Human Adipose-Derived Mesenchymal Stem Cells Differentiation. Int J Mol Sci 2020; 21:ijms21124415. [PMID: 32575918 PMCID: PMC7352648 DOI: 10.3390/ijms21124415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Understanding the molecular basis of adipogenesis is vital to identify new therapeutic targets to improve anti-obesity drugs. The adipogenic process could be a new target in the management of this disease. Our aim was to evaluate the effect of GMG-43AC, a selective peroxisome proliferator-activated receptor γ (PPARγ) modulator, during adipose differentiation of murine pre-adipocytes and human Adipose Derived Stem Cells (hADSCs). We differentiated 3T3-L1 cells and primary hADSCs in the presence of various doses of GMG-43AC and evaluated the differentiation efficiency measuring lipid accumulation, the expression of specific differentiation markers and the quantification of accumulated triglycerides. The treatment with GMG-43AC is not toxic as shown by cell viability assessments after the treatments. Our findings demonstrate the inhibition of lipid accumulation and the significant decrease in the expression of adipocyte-specific genes, such as PPARγ, FABP-4, and leptin. This effect was long lasting, as the removal of GMG-43AC from culture medium did not allow the restoration of adipogenic process. The above actions were confirmed in hADSCs exposed to adipogenic stimuli. Together, these results indicate that GMG-43AC efficiently inhibits adipocytes differentiation in murine and human cells, suggesting its possible function in the reversal of adipogenesis and modulation of lipolysis.
Collapse
Affiliation(s)
- Ghina Al Haj
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Federica Rey
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Toniella Giallongo
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Mattia Colli
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Barbara Marzani
- Research and Development, Giuliani SpA, Via Pelagio Palagi, 2, 20129 Milan, Italy; (B.M.); (G.G.)
| | - Giammaria Giuliani
- Research and Development, Giuliani SpA, Via Pelagio Palagi, 2, 20129 Milan, Italy; (B.M.); (G.G.)
| | - Alfredo Gorio
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Gian Vicenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy;
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Anna Maria Di Giulio
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
- Correspondence: (A.M.D.G.); (S.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy;
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
- Correspondence: (A.M.D.G.); (S.C.)
| |
Collapse
|
27
|
Bisicchia S, Bernardi G, Pagnotta SM, Tudisco C. Micro-fragmented stromal-vascular fraction plus microfractures provides better clinical results than microfractures alone in symptomatic focal chondral lesions of the knee. Knee Surg Sports Traumatol Arthrosc 2020; 28:1876-1884. [PMID: 31297576 DOI: 10.1007/s00167-019-05621-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE To evaluate clinical outcomes over a 1-year period in patients affected by symptomatic focal chondral lesions of the knee treated with micro-fragmented stromal-vascular fraction plus microfractures compared to microfractures alone. METHODS Two groups of 20 patients were arthroscopically treated with microfractures for a symptomatic focal chondral defect of the knee. At the end of surgery, in the experimental group, micro-fragmented stromal-vascular fraction was injected into the joint. Primary end point was WOMAC score at 12 months. Secondary end points were any adverse events, Oxford Knee Score, EQ-5D score, VAS for pain, analgesic and anti-inflammatory consumption. RESULTS All the patients were evaluated at 12-month follow-up. No adverse reactions were noted. Analgesic and anti-inflammatory consumption was similar in both groups. At 1-month follow-up, no differences were noted between groups when compared to pre-operative scores. At 3-month follow-up, patients in both groups improved from the baseline in all variables. Significantly lower VAS scores were found in the experimental group (4.2 ± 3.2 vs. 5.9 ± 1.7, p = 0.04). At 6- and 12-month follow-ups, patients in the experimental group scored better in all outcomes with a moderate effect size; in particular, better WOMAC scores were obtained at 12 months, achieving the primary end-point of the study (17.7 ± 11.1 vs. 25.5 ± 12.7; p = 0.03). CONCLUSIONS Injection of micro-fragmented stromal-vascular fraction is safe and, when associated with microfractures, is more effective in clinical terms than microfractures alone in patients affected by symptomatic focal chondral lesions of the knee. Results of the current study provide information that could help physicians to improve their counseling for patients concerning ADMSCs. LEVEL OF EVIDENCE Level 1-therapeutic study.
Collapse
Affiliation(s)
- Salvatore Bisicchia
- Applied Biotechnologies and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy. .,Department of Orthopaedic Surgery, Sports Traumatology Unit, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy.
| | - Gabriele Bernardi
- Department of Orthopaedic Surgery, Sports Traumatology Unit, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Susanna M Pagnotta
- Department of Orthopaedic Surgery, Sports Traumatology Unit, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Cosimo Tudisco
- Department of Orthopaedic Surgery, Sports Traumatology Unit, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| |
Collapse
|
28
|
Kunze KN, Burnett RA, Wright-Chisem J, Frank RM, Chahla J. Adipose-Derived Mesenchymal Stem Cell Treatments and Available Formulations. Curr Rev Musculoskelet Med 2020; 13:264-280. [PMID: 32328959 DOI: 10.1007/s12178-020-09624-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The use of human adipose-derived mesenchymal stem cells (ADSCs) has gained attention due to its potential to expedite healing and the ease of harvesting; however, clinical evidence is limited, and questions concerning optimal method of delivery and long-term outcomes remain unanswered. RECENT FINDINGS Administration of ADSCs in animal models has been reported to aid in improved healing benefits with enhanced repair biomechanics, superior gross histological appearance of injury sites, and higher concentrations of growth factors associated with healing compared to controls. Recently, an increasing body of research has sought to examine the effects of ADSCs in humans. Several available processing techniques and formulations for ADSCs exist with evidence to suggest benefits with the use of ADSCs, but the superiority of any one method is not clear. Evidence from the most recent clinical studies available demonstrates promising outcomes following treatment of select musculoskeletal pathologies with ADSCs despite reporting variability among ADSCs harvesting and processing; these include (1) healing benefits and pain improvement for rotator cuff and Achilles tendinopathies, (2) improvements in pain and function in those with knee and hip osteoarthritis, and (3) improved cartilage regeneration for osteochondral focal defects of the knee and talus. The limitation to most of this literature is the use of other therapeutic biologics in combination with ADSCs. Additionally, many studies lack control groups, making establishment of causation inappropriate. It is imperative to perform higher-quality studies using consistent, predictable control populations and to standardize formulations of ADSCs in these trials.
Collapse
Affiliation(s)
- Kyle N Kunze
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Robert A Burnett
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Joshua Wright-Chisem
- Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Rachel M Frank
- Department of Orthopaedic Surgery, Division of Sports Medicine, University of Colorado School of Medicine, Boulder, CO, USA
| | - Jorge Chahla
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
29
|
Zhou X, Zhang F, Wang D, Wang J, Wang C, Xia K, Ying L, Huang X, Tao Y, Chen S, Xue D, Hua J, Liang C, Chen Q, Li F. Micro Fragmented Adipose Tissue Promotes the Matrix Synthesis Function of Nucleus Pulposus Cells and Regenerates Degenerated Intervertebral Disc in a Pig Model. Cell Transplant 2020; 29:963689720905798. [PMID: 32030997 PMCID: PMC7444234 DOI: 10.1177/0963689720905798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) degeneration and consequent lower back pain is a common
disease. Micro fragmented adipose tissue (MFAT) is promising for a wide range of
applications in regenerative medicine. In this study, MFAT was isolated by a
nonenzymatic method and co-cultured with nucleus pulposus cells (NPCs) using an
indirect co-culture system in vitro. A pig disc degeneration
model was used to investigate the regenerative effect of MFAT on degenerated
IVDs in vivo. The mRNA expression of Sox9,
Acan, and Col2 in NPCs was significantly
increased, while no significant increase was observed in the mRNA expression of
proinflammatory cytokine genes after the NPCs were co-cultured with MFAT.
Nucleus pulposus (NP)-specific markers were increased in MFAT cells after
co-culture with NPCs. After injection of MFAT, the disc height, water content,
extracellular matrix, and structure of the degenerated NP were significantly
improved. MFAT promoted the matrix synthesis function of NPCs, and NPCs
stimulated the NP-like differentiation of MFAT cells. In addition, MFAT also
partly regenerated degenerated IVDs in the pig model.
Collapse
Affiliation(s)
- Xiaopeng Zhou
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,These authors contributed equally to this article
| | - Feng Zhang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,These authors contributed equally to this article
| | - Dawei Wang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,These authors contributed equally to this article
| | - Jingkai Wang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,These authors contributed equally to this article
| | - Chenggui Wang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Kaishun Xia
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Liwei Ying
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xianpeng Huang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yiqing Tao
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Shouyong Chen
- Department of Orthopedics Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Deting Xue
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianming Hua
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Chengzhen Liang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Qixin Chen
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fangcai Li
- Department of Orthopedics Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
30
|
Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the Different Preparation Methods to Obtain Human Adipose-Derived Stromal Vascular Fraction Cells (AD-SVFs) and Human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs): Enzymatic Digestion Versus Mechanical Centrifugation. Int J Mol Sci 2019; 20:5471. [PMID: 31684107 PMCID: PMC6862236 DOI: 10.3390/ijms20215471] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/27/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
Autologous therapies using adipose-derived stromal vascular fraction (AD-SVFs) and adult adipose-derived mesenchymal stem cells (AD-MSCs) warrant careful preparation of the harvested adipose tissue. Currently, no standardized technique for this preparation exists. Processing quantitative standards (PQSs) define manufacturing quantitative variables (such as time, volume, and pressure). Processing qualitative standards (PQLSs) define the quality of the materials and methods in manufacturing. The purpose of the review was to use PQSs and PQLSs to report the in vivo and in vitro results obtained by different processing kits that use different procedures (enzymatic vs. non-enzymatic) to isolate human AD-SVFs/AD-MSCs. PQSs included the volume of fat tissue harvested and reagents used, the time/gravity of centrifugation, and the time, temperature, and tilt level/speed of incubation and/or centrifugation. PQLSs included the use of a collagenase, a processing time of 30 min, kit weight, transparency of the kit components, the maintenance of a closed sterile processing environment, and the use of a small centrifuge and incubating rocker. Using a kit with the PQSs and PQLSs described in this study enables the isolation of AD-MSCs that meet the consensus quality criteria. As the discovery of new critical quality attributes (CQAs) of AD-MSCs evolve with respect to purity and potency, adjustments to these benchmark PQSs and PQLs will hopefully isolate AD-MSCs of various CQAs with greater reproducibility, quality, and safety. Confirmatory studies will no doubt need to be completed.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Barbara De Angelis
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Ashutosh Kothari
- Chief of Breast Surgery Unit, Guy's Hospital, Guy's and St. Thomas' NHS Foundation Trust, London SE1 9RT, UK.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
31
|
Adipose-Derived Stem Cells from Fat Tissue of Breast Cancer Microenvironment Present Altered Adipogenic Differentiation Capabilities. Stem Cells Int 2019; 2019:1480314. [PMID: 31511776 PMCID: PMC6710814 DOI: 10.1155/2019/1480314] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells able to differentiate into multiple cell types, including adipocytes, osteoblasts, and chondrocytes. The role of adipose-derived stem cells (ADSCs) in cancers is significantly relevant. They seem to be involved in the promotion of tumour development and progression and relapse processes. For this reason, investigating the effects of breast cancer microenvironment on ADSCs is of high importance in order to understand the relationship between tumour cells and the surrounding stromal cells. With the current study, we aimed to investigate the specific characteristics of human ADSCs isolated from the adipose tissue of breast tumour patients. We compared ADSCs obtained from periumbilical fat (PF) of controls with ADSCs obtained from adipose tissue of breast cancer- (BC-) bearing patients. We analysed the surface antigens and the adipogenic differentiation ability of both ADSC populations. C/EBPδ expression was increased in PF and BC ADSCs induced to differentiate compared to the control while PPARγ and FABP4 expressions were enhanced only in PF ADSCs. Conversely, adiponectin expression was reduced in PF-differentiated ADSCs while it was slightly increased in differentiated BC ADSCs. By means of Oil Red O staining, we further observed an impaired differentiation capability of BC ADSCs. To investigate this aspect more in depth, we evaluated the effect of selective PPARγ activation and nutritional supplementation on the differentiation efficiency of BC ADSCs, noting that it was only with a strong differentiation stimuli that the process took place. Furthermore, we observed no response in BC ADSCs to the PPARγ inhibitor T0070907, showing an impaired activation of this receptor in adipose cells surrounding the breast cancer microenvironment. In conclusion, our study shows an impaired adipogenic differentiation capability in BC ADSCs. This suggests that the tumour microenvironment plays a key role in the modulation of the adipose microenvironment located in the surrounding tissue.
Collapse
|
32
|
Forneris N, Burlak C. Xenotransplantation literature update, May/June 2019. Xenotransplantation 2019; 26:e12547. [PMID: 31392783 DOI: 10.1111/xen.12547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Nicole Forneris
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Christopher Burlak
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
33
|
Bouglé A, Rocheteau P, Briand D, Hardy D, Verdonk F, Tremolada C, Hivelin M, Chrétien F. Beneficial role of adipose-derived mesenchymal stem cells from microfragmented fat in a murine model of duchenne muscular dystrophy. Muscle Nerve 2019; 60:328-335. [PMID: 31228273 DOI: 10.1002/mus.26614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION No etiologic therapy is available for Duchenne muscular dystrophy (DMD), but mesenchymal stem cells were shown to be effective in preclinical models of DMD. The objective of this study is to investigate the effect of microfragmented fat extracted on a murine model of DMD. METHODS Fat tissue was extracted from healthy human participants and injected IM into DMD mice. Histological analysis, cytokines, and force measurement were performed up to 4 weeks after injection. RESULTS Duchenne muscular dystrophy mice injected with microfragmented fat exhibited an improved muscle phenotype (decreased necrosis and fibrosis), a decrease of inflammatory cytokines, and increased strength. DISCUSSION Administration of microfragmented fat in key muscles may improve muscular phenotype in patients with DMD. Muscle Nerve, 2019.
Collapse
Affiliation(s)
- Adrien Bouglé
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Department of Anesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, Paris, France.,Assistance Publique-Hôpitaux de Paris, Paris, France.,Department of Anesthesiology and Critical Care Medicine, Institute of Cardiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Pierre Rocheteau
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Service Hospitalo-Universitaire, Centre Hospitalier Sainte Anne, Paris, France.,Laboratoire Universitaire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris, France
| | - David Briand
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France
| | - David Hardy
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France
| | - Franck Verdonk
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Department of Anesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, Paris, France.,Assistance Publique-Hôpitaux de Paris, Paris, France.,Department of Anesthesiology and Critical Care Department, Saint-Antoine Hospital, Paris, France
| | | | - Mikael Hivelin
- Assistance Publique-Hôpitaux de Paris, Paris, France.,Descartes University, Assistance Publique - Hôpitaux de Paris, Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France.,Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France
| | - Fabrice Chrétien
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Laboratoire Universitaire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris, France.,Descartes University, Assistance Publique - Hôpitaux de Paris, Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
34
|
Gentile P, Piccinno MS, Calabrese C. Characteristics and Potentiality of Human Adipose-Derived Stem Cells (hASCs) Obtained from Enzymatic Digestion of Fat Graft. Cells 2019; 8:282. [PMID: 30934588 PMCID: PMC6469026 DOI: 10.3390/cells8030282] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/08/2019] [Accepted: 03/19/2019] [Indexed: 12/18/2022] Open
Abstract
Human adipose-derived stem cells localize in the stromal-vascular portion, and can be ex vivo isolated using a combination of washing steps and enzymatic digestion. For this study, we undertook a histological evaluation of traditional fat graft compared with fat graft enriched with stromal vascular fraction cells isolated by the Celution™ system to assess the interactions between cells and adipose tissue before the breast injection. In addition, we reported on histological analyses of biopsies derived from fat grafted (traditional or enriched with SVFs) in the breast in order to assess the quality of the adipose tissue, fibrosis and vessels. The hASCs derived from enzymatic digestion were systematically characterized for growth features, phenotype and multi-potent differentiation potential. They fulfill the definition of mesenchymal stem cells, albeit with a higher neural phenotype profile. These cells also express genes that constitute the core circuitry of self-renewal such as OCT4, SOX2, NANOG and neurogenic lineage genes such as NEUROD1, PAX6 and SOX3. Such findings support the hypothesis that hASCs may have a potential usefulness in neurodegenerative conditions. These data can be helpful for the development of new therapeutic approaches in personalized medicine to assess safety and efficacy of the breast reconstruction.
Collapse
Affiliation(s)
- Pietro Gentile
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| | - Maria Serena Piccinno
- Scientific and Technological Park fo Medicine "Mario Veronesi", via 29 Maggio, 6, 41037 Mirandola, Italy.
| | - Claudio Calabrese
- The Oncologic and Reconstructive Surgery Breast Unit, Oncology Department, Careggi University Hospital, Firenze 50134, Italy.
- San Rossore Breast Unit, Pisa 56122, Italy.
| |
Collapse
|
35
|
Nishimura M, Nguyen L, Watanabe N, Fujita Y, Sawamoto O, Matsumoto S. Development and characterization of novel clinical grade neonatal porcine bone marrow-derived mesenchymal stem cells. Xenotransplantation 2019; 26:e12501. [PMID: 30768802 DOI: 10.1111/xen.12501] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/01/2019] [Accepted: 01/18/2019] [Indexed: 12/22/2022]
Abstract
Due to recent advances in research on mesenchymal stem cells (MSCs), MSCs are expected to be used in various clinical applications. However, securing adequate cadaveric donors and safety of living donors are major issues. To solve such issues, we have examined to develop clinical grade neonatal porcine bone marrow-derived MSCs (npBM-MSCs). Clinical grade neonatal porcine bone marrow cells were collected, frozen, and sent to our laboratory by air. The npBM-MSCs were isolated from thawed bone marrow cells, then frozen. The thawed npBM-MSCs were examined for CD markers and differentiated into chondrocytes, osteocytes, and adipocytes. They were compared with human bone marrow-derived MSCs (hBM-MSCs) for growth rate and size. To assess the robustness of proliferation, we compared culture medium with or without gelatin. The npBM-MSCs expressed positive MSC markers CD29, CD44, and CD90 and were differentiated into chondrocytes, osteocytes, and adipocytes. The doubling time of npBM-MSCs was significantly shorter than that of hBM-MSCs (17.3 ± 0.8 vs 62.0 ± 19.6 hours, P < 0.01). The size of npBM-MSCs was also significantly smaller than that of hBM-MSCs (13.1 ± 0.3 vs 17.5 ± 0.4 μm, P < 0.001). The npBM-MSCs showed similar proliferation characters irrespective of with or without gelatin coating. The npBM-MSCs secreted VEGF-A, VEGF-C, and TGF-β1. We have established npBM-MSCs which show super-rapid growth, small size, and robust proliferation profile. The np-MSCs might be able to solve the donor issues for MSC therapy.
Collapse
Affiliation(s)
- Masuhiro Nishimura
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan
| | - Luan Nguyen
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan.,Otsuka America Pharmaceutical, Inc. (OAPI), Schaumburg, Illinois
| | - Natsuki Watanabe
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan
| | - Yasutaka Fujita
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan
| | - Osamu Sawamoto
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan
| | - Shinichi Matsumoto
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Japan
| |
Collapse
|
36
|
Jones IA, Wilson M, Togashi R, Han B, Mircheff AK, Thomas Vangsness JR C. A randomized, controlled study to evaluate the efficacy of intra-articular, autologous adipose tissue injections for the treatment of mild-to-moderate knee osteoarthritis compared to hyaluronic acid: a study protocol. BMC Musculoskelet Disord 2018; 19:383. [PMID: 30355323 PMCID: PMC6201482 DOI: 10.1186/s12891-018-2300-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/11/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a highly debilitating joint disease that causes progressive, irreversible damage to articular cartilage. OA takes a massive toll on society that has grown in recent decades, but no therapy has been shown to halt or reverse the progression of the disease. The critical need for better treatments and increased interest cellular therapies has spawned a new generation of "minimally manipulated" cell treatments. Autologous adipose tissue injections are among the most controversial of these new treatments. Despite a lack of clinical evidence, adipose tissue injections are often marketed as "stem cell" injections with wide-ranging regenerative benefits. The purpose of this study is to estimate the effect size of the treatment by comparing the efficacy of autologous fat to hyaluronic acid (HA). As a secondary aim, we will test for preliminary evidence of efficacy of autologous fat vs. HA. METHODS This is a prospective, single-center, parallel-group, randomized, controlled trial. Participants (n = 54) will receive either a single intra-articular, ultrasound-guided injection of autologous adipose tissue or a single intra-articular, ultrasound-guided injection of HA (1:1 ratio). Outcome data will be obtained at baseline, week-6 and month-6. The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain domain (WOMAC-A) will be used as the primary outcome measure. Secondary clinical outcome measures include WOMAC (full), clinical anchors (pain, function, and stiffness), and the 29-point Patient-Reported Outcomes Measurement Information System (PROMIS®) profile. We will also take synovial fluid samples and assess sway velocity using a force plate, as well as analyze excess/discard adipose tissue to gain a better understanding of how intra-articular adipose tissue injections influence the biochemical environment of the joint. DISCUSSION Given the widespread use of intra-articular fat injections in the United States, it is critical that randomized, controlled human studies evaluating efficacy and biological activity be performed. This study is the first step in addressing this unmet need, but it is not without limitations. The most notable limitations of this study are its small size and lack of blinding, which predisposes the study to both investigator and participant bias. TRIAL REGISTRATION NCT03242707 // HS-17-00365 // Registration Date (First Posted): August 8, 2018.
Collapse
Affiliation(s)
- Ian A. Jones
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, HCT 1520 San Pablo Street, suite 2000, Los Angeles, CA 90033 USA
| | - Melissa Wilson
- Department of Preventive Medicine, Keck School of Medicine of USC, 2001 Soto Street, SSB1 318A, Los Angeles, CA 90033 USA
| | - Ryan Togashi
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, HCT 1520 San Pablo Street, suite 2000, Los Angeles, CA 90033 USA
| | - Bo Han
- Departments of Surgery and Biomedical Engineering, Keck School of Medicine of USC, 1333 San Pablo St. BMT-302, Los Angeles, CA 90033 USA
| | - Austin K. Mircheff
- Department of Physiology & Neuroscience, Keck School of Medicine of USC, 1333 San Pablo St. BMT B-11A, Los Angeles, CA 90033 USA
| | - C. Thomas Vangsness JR
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, HCT 1520 San Pablo Street, suite 2000, Los Angeles, CA 90033 USA
| |
Collapse
|
37
|
Paolella F, Manferdini C, Gabusi E, Gambari L, Filardo G, Kon E, Mariani E, Lisignoli G. Effect of microfragmented adipose tissue on osteoarthritic synovial macrophage factors. J Cell Physiol 2018; 234:5044-5055. [DOI: 10.1002/jcp.27307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/01/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Francesca Paolella
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Cristina Manferdini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Elena Gabusi
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | | | - Elizaveta Kon
- Department of Biomedical Sciences Humanitas University Milan Italy
- Humanitas Clinical and Research Center Milan Italy
| | - Erminia Mariani
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
- DIMEC, Alma Mater Studiorum, Università di Bologna Bologna Italy
| | - Gina Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| |
Collapse
|
38
|
Carelli S, Colli M, Vinci V, Caviggioli F, Klinger M, Gorio A. Mechanical Activation of Adipose Tissue and Derived Mesenchymal Stem Cells: Novel Anti-Inflammatory Properties. Int J Mol Sci 2018; 19:ijms19010267. [PMID: 29337886 PMCID: PMC5796213 DOI: 10.3390/ijms19010267] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 12/29/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The adipose tissue is a source of inflammatory proteins, such as TNF, IL-6, and CXCL8. Most of their production occurs in macrophages that act as scavengers of dying adipocytes. The application of an orbital mechanical force for 6-10 min at 97 g to the adipose tissue, lipoaspirated and treated according to Coleman procedures, abolishes the expression of TNF-α and stimulates the expression of the anti-inflammatory protein TNF-stimulated gene-6 (TSG-6). This protein had protective and anti-inflammatory effects when applied to animal models of rheumatic diseases. We examined biopsy, lipoaspirate, and mechanically activated fat and observed that in addition to the increased TSG-6, Sox2, Nanog, and Oct4 were also strongly augmented by mechanical activation, suggesting an effect on stromal cell stemness. Human adipose tissue-derived mesenchymal stem cells (hADSCs), produced from activated fat, grow and differentiate normally with proper cell surface markers and chromosomal integrity, but their anti-inflammatory action is far superior compared to those mesenchymal stem cells (MSCs) obtained from lipoaspirate. The expression and release of inflammatory cytokines from THP-1 cells was totally abolished in mechanically activated adipose tissue-derived hADSCs. In conclusion, we report that the orbital shaking of adipose tissue enhances its anti-inflammatory properties, and derived MSCs maintain such enhanced activity.
Collapse
Affiliation(s)
- Stephana Carelli
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", University of Milan, 20142 Milan, Italy.
| | - Mattia Colli
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", University of Milan, 20142 Milan, Italy.
| | - Valeriano Vinci
- Humanitas Research Hospital, Plastic Surgery Unit, Via Manzoni 56, 20089 Rozzano, Italy.
| | - Fabio Caviggioli
- Multimedica San Giuseppe Hospital, Plastic Surgery Unit, Via San Vittore 12, 20123 Milan, Italy.
| | - Marco Klinger
- Humanitas Research Hospital, Plastic Surgery Unit, Via Manzoni 56, 20089 Rozzano, Italy.
| | - Alfredo Gorio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Via A. di Rudinì 8, 20142 Milan, Italy.
| |
Collapse
|
39
|
Striano RD, Battista V, Bilboo N. Non-Responding Knee Pain with Osteoarthritis, Meniscus and Ligament Tears Treated with Ultrasound Guided Autologous, Micro-Fragmented and Minimally Manipulated Adipose Tissue. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/ojrm.2017.62002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Ceserani V, Ferri A, Berenzi A, Benetti A, Ciusani E, Pascucci L, Bazzucchi C, Coccè V, Bonomi A, Pessina A, Ghezzi E, Zeira O, Ceccarelli P, Versari S, Tremolada C, Alessandri G. Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue and its derived mesenchymal stromal cells. Vasc Cell 2016; 8:3. [PMID: 27547374 PMCID: PMC4991117 DOI: 10.1186/s13221-016-0037-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for advanced cell-based therapies. They are routinely obtained enzymatically from fat lipoaspirate (LP) as SVF, and may undergo prolonged ex vivo expansion, with significant senescence and decline in multipotency. Besides, these techniques have complex regulatory issues, thus incurring in the compelling requirements of GMP guidelines. Hence, availability of a minimally manipulated, autologous adipose tissue would have remarkable biomedical and clinical relevance. For this reason, a new device, named Lipogems® (LG), has been developed. This ready-to-use adipose tissue cell derivate has been shown to have in vivo efficacy upon transplantation for ischemic and inflammatory diseases. To broaden our knowledge, we here investigated the angiogenic and anti-inflammatory properties of LG and its derived MSC (LG-MSCs) population. METHODS Human LG samples and their LG-MSCs were analyzed by immunohistochemistry for pericyte, endothelial and mesenchymal stromal cell marker expression. Angiogenesis was investigated testing the conditioned media (CM) of LG (LG-CM) and LG-MSCs (LG-MSCs-CM) on cultured endothelial cells (HUVECs), evaluating proliferation, cord formation, and the expression of the adhesion molecules (AM) VCAM-1 and ICAM-1. The macrophage cell line U937 was used to evaluate the anti-inflammatory properties, such as migration, adhesion on HUVECs, and release of RANTES and MCP-1. RESULTS Our results indicate that LG contained a very high number of mesenchymal cells expressing NG2 and CD146 (both pericyte markers) together with an abundant microvascular endothelial cell (mEC) population. Substantially, both LG-CM and LG-MSC-CM increased cord formation, inhibited endothelial ICAM-1 and VCAM-1 expression following TNFα stimulation, and slightly improved HUVEC proliferation. The addition of LG-CM and LG-MSC-CM strongly inhibited U937 migration upon stimulation with the chemokine MCP-1, reduced their adhesion on HUVECs and significantly suppressed the release of RANTES and MCP-1. CONCLUSIONS Our data indicate that LG micro-fragmented adipose tissue retains either per se, or in its embedded MSCs content, the capacity to induce vascular stabilization and to inhibit several macrophage functions involved in inflammation.
Collapse
Affiliation(s)
- Valentina Ceserani
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| | - Anna Ferri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| | - Angiola Berenzi
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Anna Benetti
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Cinzia Bazzucchi
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Erica Ghezzi
- San Michele Veterinary Hospital, Tavezzano con Villavesco, Lodi, Italy
| | - Offer Zeira
- San Michele Veterinary Hospital, Tavezzano con Villavesco, Lodi, Italy
| | - Piero Ceccarelli
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | | | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Via Celoria 11, 20131 Milan, Italy
| |
Collapse
|
41
|
Marfia G, Navone SE, Hadi LA, Paroni M, Berno V, Beretta M, Gualtierotti R, Ingegnoli F, Levi V, Miozzo M, Geginat J, Fassina L, Rampini P, Tremolada C, Riboni L, Campanella R. The Adipose Mesenchymal Stem Cell Secretome Inhibits Inflammatory Responses of Microglia: Evidence for an Involvement of Sphingosine-1-Phosphate Signalling. Stem Cells Dev 2016; 25:1095-107. [DOI: 10.1089/scd.2015.0268] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, Milan, Italy
| | - Moira Paroni
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi,” Milan, Italy
| | - Valeria Berno
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi,” Milan, Italy
| | - Matteo Beretta
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | | | - Vincenzo Levi
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Monica Miozzo
- Division of Pathology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Jens Geginat
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi,” Milan, Italy
| | - Lorenzo Fassina
- Department of Health Sciences and Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Paolo Rampini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
42
|
De Francesco F, Romano M, Zarantonello L, Ruffolo C, Neri D, Bassi N, Giordano A, Zanus G, Ferraro GA, Cillo U. The role of adipose stem cells in inflammatory bowel disease: From biology to novel therapeutic strategies. Cancer Biol Ther 2016; 17:889-98. [PMID: 27414952 DOI: 10.1080/15384047.2016.1210741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases are an increasing phenomenon in western countries and in growing populations. The physiopathology of these conditions is linked to intestinal stem cells homeostasis and regenerative potential in a chronic inflammatory microenvironment. Patients with IBD present an increased risk of developing colorectal cancer (CRC), or colitis associated cancer (CAC). Conventional treatment for IBD target the inflammatory process (and include anti-inflammatory and immunosuppressive drugs) with biological agents emerging as a therapeutic approach for non-responders to traditional therapy. Conventional treatment provides scarce results and present severe complications. The intestinal environment may host incoming stem cells, able to engraft in the epithelial damaged sites and differentiate. Therefore, stem cell therapies represent an emerging alternative in inflammatory bowel diseases, with current investigations on the use of haematopoietic and mesenchymal stem cells, in particular adipose stem cells, apparently fundamental as regenerators and as immune-modulators. Here, we discuss stem cells in intestinal homeostasis and as therapeutic agents for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Francesco De Francesco
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Maurizio Romano
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Laura Zarantonello
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Cesare Ruffolo
- c Department of Surgery , Regional Center for hpb surgery, Regional Hospital of Treviso , TV , Italy
| | - Daniele Neri
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Nicolò Bassi
- c Department of Surgery , Regional Center for hpb surgery, Regional Hospital of Treviso , TV , Italy
| | - Antonio Giordano
- d Department of Medicine , Surgery and Neuroscience, University of Siena , Siena , Italy.,e Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia , PA , USA
| | - Giacomo Zanus
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Giuseppe A Ferraro
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Umberto Cillo
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| |
Collapse
|
43
|
Tremolada C, Colombo V, Ventura C. Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Technology Development. CURRENT STEM CELL REPORTS 2016; 2:304-312. [PMID: 27547712 PMCID: PMC4972861 DOI: 10.1007/s40778-016-0053-5] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past few years, interest in adipose tissue as an ideal source of mesenchymal stem cells (MSCs) has increased. These cells are multipotent and may differentiate in vitro into several cellular lineages, such as adipocytes, chondrocytes, osteoblasts, and myoblasts. In addition, they secrete many bioactive molecules and thus are considered "mini-drugstores." MSCs are being used increasingly for many clinical applications, such as orthopedic, plastic, and reconstructive surgery. Adipose-derived MSCs are routinely obtained enzymatically from fat lipoaspirate as SVF and/or may undergo prolonged ex vivo expansion, with significant senescence and a decrease in multipotency, leading to unsatisfactory clinical results. Moreover, these techniques are hampered by complex regulatory issues. Therefore, an innovative technique (Lipogems®; Lipogems International SpA, Milan, Italy) was developed to obtain microfragmented adipose tissue with an intact stromal vascular niche and MSCs with a high regenerative capacity. The Lipogems® technology, patented in 2010 and clinically available since 2013, is an easy-to-use system designed to harvest, process, and inject refined fat tissue and is characterized by optimal handling ability and a great regenerative potential based on adipose-derived MSCs. In this novel technology, the adipose tissue is washed, emulsified, and rinsed and adipose cluster dimensions gradually are reduced to about 0.3 to 0.8 mm. In the resulting Lipogems® product, pericytes are retained within an intact stromal vascular niche and are ready to interact with the recipient tissue after transplantation, thereby becoming MSCs and starting the regenerative process. Lipogems® has been used in more than 7000 patients worldwide in aesthetic medicine and surgery, as well as in orthopedic and general surgery, with remarkable and promising results and seemingly no drawbacks. Now, several clinical trials are under way to support the initial encouraging outcomes. Lipogems® technology is emerging as a valid intraoperative system to obtain an optimal final product that may be used immediately for regenerative purposes.
Collapse
Affiliation(s)
| | | | - Carlo Ventura
- Stem Wave Institute for Tissue Healing (SWITH)—Ettore Sansavini Health Science Foundation, Lugo, Ravenna, Italy
| |
Collapse
|
44
|
Dehkordi MB, Madjd Z, Chaleshtori MH, Meshkani R, Nikfarjam L, Kajbafzadeh AM. A Simple, Rapid, and Efficient Method for Isolating Mesenchymal Stem Cells from the Entire Umbilical Cord. Cell Transplant 2016; 25:1287-1297. [DOI: 10.3727/096368915x582769] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Several reports have been published on the isolation, culture, and identification of mesenchymal stem cells (MSCs) from different anatomical regions of the umbilical cord (UC). UC is suitable for standardizing methods of MSC isolation because it is a uniform source with high MSC numbers. Although the UC is considered a medical waste after childbirth, ethical issues for its use must be considered. An increased demand for MSCs in regenerative medicine has made scientists prioritize the development of MSC isolation methods. Several research groups are attempting to provide a large number of high-quality MSCs. In this study, we present a modulated explant/enzyme method (MEEM) to isolate the maximum number of MSCs from the entire UC. This method was established for the isolation of MSCs from different anatomical regions of the UC altogether. We could retrieve 6 to 10 million MSCs during 8 to 10 days of primary culture. After three passages, we could obtain 8–10 × 108 cells in 28–30 days. MSCs isolated by this method express CD73, CD90, CD105, and CD44, but they do not express hematopoietic markers CD34 and CD45 or the endothelial marker CD31. The genes SOX2, OCT4, and NANOG are expressed in isolated MSCs. The capacity of these MSCs to differentiate into adipocytes and osteocytes highlights their application in regenerative medicine. This method is simple, reproducible, and cost efficient. Moreover, this method is suitable for the production of a large number of high-quality MSCs from an UC in less than a month, to be used for cell therapy in an 80-kg person.
Collapse
Affiliation(s)
- Mehdi Banitalebi Dehkordi
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran (IRI)
| | - Zahra Madjd
- Oncopathology Research Center and Dep Pathology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran (IRI)
| | | | - Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Laleh Nikfarjam
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| |
Collapse
|
45
|
Latorre E, Carelli S, Caremoli F, Giallongo T, Colli M, Canazza A, Provenzani A, Di Giulio AM, Gorio A. Human Antigen R Binding and Regulation of SOX2 mRNA in Human Mesenchymal Stem Cells. Mol Pharmacol 2016; 89:243-52. [PMID: 26677051 DOI: 10.1124/mol.115.100701] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/11/2015] [Indexed: 02/02/2023] Open
Abstract
Since 2005, sex determining region y-box 2 (SOX2) has drawn the attention of the scientific community for being one of the key transcription factors responsible for pluripotency induction in somatic stem cells. Our research investigated the turnover regulation of SOX2 mRNA in human adipose-derived stem cells, considered one of the most valuable sources of somatic stem cells in regenerative medicine. Mitoxantrone is a drug that acts on nucleic acids primarily used to treat certain types of cancer and was recently shown to ameliorate the outcome of autoimmune diseases such as multiple sclerosis. In addition, mitoxantrone has been shown to inhibit the binding of human antigen R (HuR) RNA-binding protein to tumor necrosis factor-α mRNA. Our results show that HuR binds to the 3'-untranslated region of SOX2 mRNA together with the RNA-induced silencing complex miR145. The HuR binding works by stabilizing the interaction between the 3'-untranslated region and the RNA-induced silencing complex. Cell exposure to mitoxantrone leads to HuR detachment and the subsequent prolongation of the SOX2 mRNA half-life. The prolonged SOX2 half-life allows improvement of the spheroid-forming capability of the adipose-derived stem cells. The silencing of HuR confirmed the above observations and illustrates how the RNA-binding protein HuR may be a required molecule for regulation of SOX2 mRNA decay.
Collapse
Affiliation(s)
- Elisa Latorre
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Stephana Carelli
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Filippo Caremoli
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Toniella Giallongo
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Mattia Colli
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Alessandra Canazza
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Alessandro Provenzani
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Anna Maria Di Giulio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| | - Alfredo Gorio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy (E.L., S.C., F.C., T.G., M.C., A.M.D.G., A.G.); Laboratory of Cell Biology, Cerebrovascular Diseases Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy (A.C.); and Laboratory of Genomic Screening Center for Integrative Biology, University of Trento, Trento, Italy (A.P.)
| |
Collapse
|
46
|
Tremolada C, Ricordi C, Caplan AI, Ventura C. Mesenchymal Stem Cells in Lipogems, a Reverse Story: from Clinical Practice to Basic Science. Methods Mol Biol 2016; 1416:109-122. [PMID: 27236668 DOI: 10.1007/978-1-4939-3584-0_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The idea that basic science should be the starting point for modern clinical approaches has been consolidated over the years, and emerged as the cornerstone of Molecular Medicine. Nevertheless, there is increasing concern over the low efficiency and inherent costs related to the translation of achievements from the bench to the bedside. These burdens are also perceived with respect to the effectiveness of translating basic discoveries in stem cell biology to the newly developing field of advanced cell therapy or Regenerative Medicine. As an alternative paradigm, past and recent history in Medical Science provides remarkable reverse stories in which clinical observations at the patient's bedside have fed major advances in basic research which, in turn, led to consistent progression in clinical practice. Within this context, we discuss our recently developed method and device, which forms the core of a system (Lipogems) for processing of human adipose tissue solely with the aid of mild mechanical forces to yield a microfractured tissue product.
Collapse
Affiliation(s)
| | - Camillo Ricordi
- Cell Transplant Program and Diabetes Research Institute, University of Miami, Miami, FL, USA
| | - Arnold I Caplan
- Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Carlo Ventura
- SWITH (Stem Wave Institute for Tissue Healing), Gruppo Villa Maria (GVM) and Ettore Sansavini Health Science Foundation - ONLUS, Lugo (Ravenna), Italy.
- National Institute of Biostructures and Biosystems (NIBB) at the S. Orsola - Malpighi Hospital, Institute of Cardiology, University of Bologna, Pavilion 21, Via Massarenti N. 9, 40138, Bologna, Italy.
| |
Collapse
|
47
|
Mesenchymal Stem/Stromal Cells from Discarded Neonatal Sternal Tissue: In Vitro Characterization and Angiogenic Properties. Stem Cells Int 2015; 2016:5098747. [PMID: 26770206 PMCID: PMC4684890 DOI: 10.1155/2016/5098747] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
Autologous and nonautologous bone marrow mesenchymal stem/stromal cells (MSCs) are being evaluated as proangiogenic agents for ischemic and vascular disease in adults but not in children. A significant number of newborns and infants with critical congenital heart disease who undergo cardiac surgery already have or are at risk of developing conditions related to inadequate tissue perfusion. During neonatal cardiac surgery, a small amount of sternal tissue is usually discarded. Here we demonstrate that MSCs can be isolated from human neonatal sternal tissue using a nonenzymatic explant culture method. Neonatal sternal bone MSCs (sbMSCs) were clonogenic, had a surface marker expression profile that was characteristic of bone marrow MSCs, were multipotent, and expressed pluripotency-related genes at low levels. Neonatal sbMSCs also demonstrated in vitro proangiogenic properties. Sternal bone MSCs cooperated with human umbilical vein endothelial cells (HUVECs) to form 3D networks and tubes in vitro. Conditioned media from sbMSCs cultured in hypoxia also promoted HUVEC survival and migration. Given the neonatal source, ease of isolation, and proangiogenic properties, sbMSCs may have relevance to therapeutic applications.
Collapse
|
48
|
Oberbauer E, Steffenhagen C, Wurzer C, Gabriel C, Redl H, Wolbank S. Enzymatic and non-enzymatic isolation systems for adipose tissue-derived cells: current state of the art. CELL REGENERATION (LONDON, ENGLAND) 2015; 4:7. [PMID: 26435835 PMCID: PMC4591586 DOI: 10.1186/s13619-015-0020-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/23/2015] [Indexed: 02/07/2023]
Abstract
In the past decade, adipose tissue became a highly interesting source of adult stem cells for plastic surgery and regenerative medicine. The isolated stromal vascular fraction (SVF) is a heterogeneous cell population including the adipose-derived stromal/stem cells (ASC), which showed regenerative potential in several clinical studies and trials. SVF should be provided in a safe and reproducible manner in accordance with current good manufacturing practices (cGMP). To ensure highest possible safety for patients, a precisely defined procedure with a high-quality control is required. Hence, an increasing number of adipose tissue-derived cell isolation systems have been developed. These systems aim for a closed, sterile, and safe isolation process limiting donor variations, risk for contaminations, and unpredictability of the cell material. To isolate SVF from adipose tissue, enzymes such as collagenase are used. Alternatively, in order to avoid enzymes, isolation systems using physical forces are available. Here, we provide an overview of known existing enzymatic and non-enzymatic adipose tissue-derived cell isolation systems, which are patented, published, or already on the market.
Collapse
Affiliation(s)
- Eleni Oberbauer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Carolin Steffenhagen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christoph Wurzer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christian Gabriel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz/Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
49
|
Wang S, Mundada L, Johnson S, Wong J, Witt R, Ohye RG, Si MS. Characterization and angiogenic potential of human neonatal and infant thymus mesenchymal stromal cells. Stem Cells Transl Med 2015; 4:339-50. [PMID: 25713463 DOI: 10.5966/sctm.2014-0240] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Resident mesenchymal stromal cells (MSCs) are involved in angiogenesis during thymus regeneration. We have previously shown that MSCs can be isolated from enzymatically digested human neonatal and infant thymus tissue that is normally discarded during pediatric cardiac surgical procedures. In this paper, we demonstrate that thymus MSCs can also be isolated by explant culture of discarded thymus tissue and that these cells share many of the characteristics of bone marrow MSCs. Human neonatal thymus MSCs are clonogenic, demonstrate exponential growth in nearly 30 population doublings, have a characteristic surface marker profile, and express pluripotency genes. Furthermore, thymus MSCs have potent proangiogenic behavior in vitro with sprout formation and angiogenic growth factor production. Thymus MSCs promote neoangiogenesis and cooperate with endothelial cells to form functional human blood vessels in vivo. These characteristics make thymus MSCs a potential candidate for use as an angiogenic cell therapeutic agent and for vascularizing engineered tissues in vitro.
Collapse
Affiliation(s)
- Shuyun Wang
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lakshmi Mundada
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sean Johnson
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Joshua Wong
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Russell Witt
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Richard G Ohye
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ming-Sing Si
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery and Department of Pediatric Cardiology, University of Michigan, Ann Arbor, Michigan, USA; Department of General Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Hopkins AM, DeSimone E, Chwalek K, Kaplan DL. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125:1-25. [PMID: 25461688 PMCID: PMC4324093 DOI: 10.1016/j.pneurobio.2014.11.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/12/2014] [Accepted: 11/15/2014] [Indexed: 12/15/2022]
Abstract
There are currently more than 600 diseases characterized as affecting the central nervous system (CNS) which inflict neural damage. Unfortunately, few of these conditions have effective treatments available. Although significant efforts have been put into developing new therapeutics, drugs which were promising in the developmental phase have high attrition rates in late stage clinical trials. These failures could be circumvented if current 2D in vitro and in vivo models were improved. 3D, tissue-engineered in vitro systems can address this need and enhance clinical translation through two approaches: (1) bottom-up, and (2) top-down (developmental/regenerative) strategies to reproduce the structure and function of human tissues. Critical challenges remain including biomaterials capable of matching the mechanical properties and extracellular matrix (ECM) composition of neural tissues, compartmentalized scaffolds that support heterogeneous tissue architectures reflective of brain organization and structure, and robust functional assays for in vitro tissue validation. The unique design parameters defined by the complex physiology of the CNS for construction and validation of 3D in vitro neural systems are reviewed here.
Collapse
Affiliation(s)
- Amy M Hopkins
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Elise DeSimone
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Karolina Chwalek
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|