1
|
Wei Y, Xie C, Wei Y, Li Z, Li L, Chen Y, Jia C, Xie H, Liao J. SVF Cell Sheets as a New Multicellular Material-Based Strategy for Promoting Angiogenesis and Regeneration in Diced Cartilage Grafts. J Craniofac Surg 2025:00001665-990000000-02621. [PMID: 40209026 DOI: 10.1097/scs.0000000000011358] [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: 11/23/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025] Open
Abstract
Autologous diced cartilage, while biocompatible and easy to shape, is limited in clinical application due to its high adsorption rate and challenges in establishing timely and effective neovascularization postsurgery. In this study, the authors produced SVF cell sheets from adipose-derived stromal vascular fraction (SVF) through enzymatic digestion, employing a temperature-sensitive culture system. Our in vivo and in vitro experiments validated that SVF cell sheets, when wrapped around granular cartilage, exhibited a notable promotion of cartilage regeneration and mitigated granular cartilage adsorption in a rabbit diced cartilage graft model. Our findings demonstrate that SVF cell sheets facilitated effective neovascularization and timely cartilage block formation by secreting VEGF and Ang-1 while also suppressing the expression of pyroptotic proteins like NLRP3, Caspase1, and GSDMD. As a biofilm, derived from a multicellular source, SVF cell sheets can replace perichondrium and promote the expression of proangiogenic growth factors Ang-1 and VEGF, thereby promoting local microvascular regeneration, reducing chondrocyte pyroptosis, and promoting the formation of cartilage blocks. This strategy provides a potential new method for autologous cartilage grafting, which will help solve the dilemma of limited sources of cartilage tissue in clinical practice and provide natural autologous cartilage filling materials for the treatment of craniofacial defects.
Collapse
Affiliation(s)
- Yangchen Wei
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Cong Xie
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Yi Wei
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
- Department of Plastic and Reconstructive Surgery, Central South University Third Xiangya Hospital
| | - Zhengyang Li
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Li Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Yan Chen
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Chiyu Jia
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| | - Hongju Xie
- Department of Plastic Surgery, The Second Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Junlin Liao
- Center of Burn and Plastic and Wound Repair, The First Affiliated Hospital, Hengyang Medical School, University of South China
| |
Collapse
|
2
|
Wiest EF, Zubair AC. Generation of Current Good Manufacturing Practices-Grade Mesenchymal Stromal Cell-Derived Extracellular Vesicles Using Automated Bioreactors. BIOLOGY 2025; 14:313. [PMID: 40136569 PMCID: PMC11940689 DOI: 10.3390/biology14030313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Interest in Current Good Manufacturing Practices (cGMP)-grade extracellular vesicles (EVs) is expanding. Some obstacles in this new but rapidly growing field include a lack of standardization and scalability. This review focuses on automated biomanufacturing of EVs in conditioned media collected from cultured mesenchymal stromal cells (MSCs). Different automated cell culture systems are discussed, including factors affecting EV quantity and quality, isolating EVs manufactured in an automated system, and validations needed. The ultimate goal when manufacturing cGMP-grade EVs is to identify a specific application and characterize the EV population in detail. This is achieved by validating every step of the process, choosing appropriate release criteria, and assuring batch-to-batch consistency. Due to the lack of standards in the field, it is critical to ensure that the cGMP-grade EVs meet FDA standards pertaining to identity, reproducibility, sterility, safety, purity, and potency. A closed-system automated bioreactor can be a valuable tool to generate cGMP-EVs in a scalable, economical, and reproducible manner.
Collapse
Affiliation(s)
- Elani F. Wiest
- Department of Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Abba C. Zubair
- Department of Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL 32224, USA;
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
3
|
Fasano A, Matteoli M. Just a fat joke? Time will tell. Parkinsonism Relat Disord 2025; 132:107298. [PMID: 39880739 DOI: 10.1016/j.parkreldis.2025.107298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Affiliation(s)
- Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, Ontario, Canada; Division of Neurology, University of Toronto, Toronto, Ontario, Canada; Krembil Brain Institute, Toronto, Ontario, Canada; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy.
| | - Michela Matteoli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| |
Collapse
|
4
|
Fang Y, Gonzales-Nieves S, Cifarelli V, Imoukhuede PI. Sex Differences in VEGF and PDGF Ligand and Receptor Protein Expression during Adipose Tissue Expansion and Regression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638177. [PMID: 40027606 PMCID: PMC11870404 DOI: 10.1101/2025.02.14.638177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Inadequate angiogenesis in obesogenic adipose tissue (AT) has been implicated in disrupted adipogenesis and metabolic disorders. Yet, key cellular and molecular regulators of AT angiogenesis remain largely unidentified. This study sought to identify the dysregulated elements within the Vascular Endothelial Growth Factor (VEGF) and Platelet-Derived Growth Factor (PDGF) systems during obesity progression. We employ a mouse model, comprising both male and female mice, to investigate the changes in the VEGF/PDGF concentration and their receptor distribution in AT during short- and long-term weight gain and weight loss. Our results reveal pronounced sex-specific differences in obesity progression, with male and female mice exhibiting distinct angiogenic ligand and receptor profiles under identical dietary interventions. This data also lays the groundwork for developing computational models of VEGF/PDGF signaling networks in AT, allowing for the simulation of complex biological interactions and the prediction of therapeutic outcomes.
Collapse
|
5
|
Fish SR, Halley CL, Dileepan M, Hertzel AV, Dickey DM, Bernlohr DA. Expression of fatty acid binding proteins in mesenteric adipose tissue. Biochem Biophys Res Commun 2025; 749:151346. [PMID: 39855040 DOI: 10.1016/j.bbrc.2025.151346] [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: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Adipose is a complex tissue comprised of adipocytes, immune cells, endothelial and progenitor stem cells. In humans, there are at least nine defined adipose depots, each containing variable numbers of genetically identified adipocyte clusters suggesting remarkable heterogeneity and potential functionality in each depot with respect to lipid metabolism. Although subcutaneous and visceral depots are commonly analyzed for biochemical and molecular functions, the mesenteric depot has been overlooked yet strongly implicated in lipid mediated immune surveillance. Since fatty acid binding proteins (FABPs) are primary cellular conduits to lipid trafficking, we evaluated the expression patterns for four major fatty acid binding proteins (FABP1, FABP3, FABP4 and FABP5) using a combination of gene expression, immunoblotting, and immunofluorescence in mesenteric fat from both young and old, male and female C57Bl/6J mice. All four FABPs were expressed at the mRNA and protein level in murine mesenteric adipose tissue. While there was no statistical change in expression of mesenteric FABP isoforms with sex or age, the expression of mesenteric FABP1 was increased, and FABP4 decreased, in both males and females as compared to perigonadal and inguinal depots. Surprisingly, immunofluorescence staining revealed that compared to subcutaneous or perigonadal depots, mesenteric fat expresses FABP3, but little FABP5, in adipocytes. These results highlight the diversity in adipose tissue and the importance of evaluating the mesenteric depot in the context of lipid transport and metabolism.
Collapse
Affiliation(s)
- Shayla R Fish
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Catherine L Halley
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Mythili Dileepan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA; Institute on the Biology of Aging and Metabolism, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA; Institute on the Biology of Aging and Metabolism, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Deborah M Dickey
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, USA; Institute on the Biology of Aging and Metabolism, University of Minnesota-Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
6
|
Chieng CY, Mannapperuma N, Sayan A, Ilankovan V. Actinic cheilitis: Do stem cells have a role in its management? A case series. Br J Oral Maxillofac Surg 2025:S0266-4356(25)00020-8. [PMID: 40118757 DOI: 10.1016/j.bjoms.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 03/23/2025]
Abstract
Actinic cheilitis (AC) is a premalignant mucosal lip condition that mainly affects the lower lip and has a high propensity of malignant transformation into squamous cell carcinoma. There is no gold standard for its management, but early intervention aims to minimise the risk of malignant transformation. Here we report our experience of utilising nanofat grafting in the management of AC. Retrospective data analysis was carried out between 2020 and 2024. Patients diagnosed with AC were treated with nanofat grafting. The nanofat preparation was harvested from the abdomen and prepared using the PureGraft® filtration system (Bimini Health Tech), and was infiltrated into the lips in two layers. Outcomes were assessed clinically. Seven patients were included in our analysis with a mean follow up of 23 months. All patients had had previous treatment for AC such as topical 5-fluororacil, steroids, laser ablation, and cryotherapy. Five of the seven received simultaneous nanofat grafting followed by erbium laser resurfacing at the same appointment. The others had nanofat grafting after laser resurfacing. Following the procedure all the patients had no further cracks, ulcerations, or erosions of the lips, with a good definition of the vermilion border, and all patients remained symptom free. The combination of nanofat grafting and laser resurfacing provides an alternative minimally invasive solution for patients with persistent AC. We have formulated an algorithm for management based on our experience.
Collapse
Affiliation(s)
- C Y Chieng
- Medical Student, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom.
| | - N Mannapperuma
- Clinical Fellow in Oral & Maxillofacial Surgery, University Hospitals Dorset NHS Trust, United Kingdom
| | - A Sayan
- Specialty Registrar in Oral & Maxillofacial Surgery, Oxford University Hospital NHS Foundation Trust, United Kingdom
| | - V Ilankovan
- Consultant in Oral & Maxillofacial Surgery, Poole Hospital, University Hospital Dorset NHS Trust, United Kingdom
| |
Collapse
|
7
|
Lee HY, An SB, Hwang SY, Hwang GY, Lee HL, Park HJ, Shin J, Kim KN, Wee SW, Yoon SL, Ha Y. Synergistic enhancement of spinal fusion in preclinical models using low-dose rhBMP-2 and stromal vascular fraction in an injectable hydrogel composite. Mater Today Bio 2025; 30:101379. [PMID: 39759847 PMCID: PMC11699625 DOI: 10.1016/j.mtbio.2024.101379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Spinal fusion surgery remains a significant challenge due to limitations in current bone graft materials, particularly in terms of bioactivity, integration, and safety. This study presents an innovative approach using an injectable hydroxyapatite/β-tricalcium phosphate (HA/β-TCP) hydrogel combined with stromal vascular fraction (SVF) and low-dose recombinant human BMP-2 (rhBMP-2) to enhance osteodifferentiation and angiogenesis. Through a series of in vitro studies and preclinical models involving rats and minipigs, we demonstrated that the hydrogel system enables the sustained release of rhBMP-2, resulting in significantly improved bone density and integration, alongside reduced inflammatory responses. The combination of rhBMP-2 and SVF in this injectable formulation yielded superior spinal fusion outcomes, with enhanced mechanical properties and increased bone mass in both small and large animal models. These findings suggest that this strategy offers a promising and safer alternative for spinal fusion, with strong potential for clinical application.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Graduate School, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Jung Park
- Department of Research Center, CGBio., co. Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Joongkyum Shin
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung Won Wee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sol Lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| |
Collapse
|
8
|
Lee Y, Tukei KL, Fang Y, Kuila S, Liu X, Imoukhuede PI. Integrative analysis of angiogenic signaling in obesity: capillary features and VEGF binding kinetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630107. [PMID: 39763822 PMCID: PMC11703262 DOI: 10.1101/2024.12.23.630107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Obesity is a global health crisis, with its prevalence particularly severe in the United States, where over 42% of adults are classified as obese. Obesity is driven by complex molecular and tissue-level mechanisms that remain poorly understood. Among these, angiogenesis-primarily mediated by vascular endothelial growth factor (VEGF-A)-is critical for adipose tissue expansion but presents unique challenges for therapeutic targeting due to its intricate regulation. Systems biology approaches have advanced our understanding of VEGF-A signaling in vascular diseases, but their application to obesity is limited by scattered and sometimes contradictory data. To address this gap, we performed a comprehensive analysis of the existing literature to synthesize key findings, standardize data, and provide a holistic perspective on the adipose vascular microenvironment. The data mining revealed five key findings: (1) obesity increases adipocyte size by 78%; (2) vessel density in adipose tissue decreases by 51% in obese mice, with vessels being 47-58% smaller and 4-9 times denser in comparison with tumor vessels; (3) capillary basement membrane thickness remains similar regardless of obesity; (4) VEGF-A shows the strongest binding affinity for VEGFR1, with four times stronger affinity for VEGFR2 than for NRP1; and (5) binding affinities measured by radioligand binding assay and surface plasmon resonance (SPR) are significantly different. These consolidated findings provide essential parameters for systems biology modeling, new insights into obesity-induced changes in adipose tissue, and a foundation for developing angiogenesis-targeting therapies for obesity.
Collapse
|
9
|
Ramakrishnan VM, Thaker H, Ocampo GL, Adam RM, Estrada CR. Pediatric bladder tissue engineering: Where have we been and where do we go next? J Pediatr Urol 2025:S1477-5131(25)00001-4. [PMID: 39827049 DOI: 10.1016/j.jpurol.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/11/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES This review aims to (a) provide a concise overview of early clinical trials in bladder tissue engineering and the associated challenges, (b) evaluate significant advancements over the past 15 years in addressing key limitations in angiogenesis, scaffolding, cell sourcing, and immunomodulation, and (c) explore the individual and synergistic contributions of each domain toward the development of a viable engineered solution. MATERIALS AND METHODS Relevant papers for this narrative review were selected through a PubMed search for "bladder tissue engineering" studies published between 01/01/2009 and 12/31/2024, as well as earlier clinical trials that predate this period. RESULTS Along with reviewing four major clinical trials, this review highlights nearly 20 distinct studies that showcase progress in the critical domains of angiogenesis, scaffolding, cell sourcing, and immunomodulation. CONCLUSIONS Are we close to developing an off-the-shelf bladder substitute? Not yet. Current efforts are focused on addressing two major knowledge gaps: (a) the lack of testing in clinically relevant disease models and (b) the need for a more comprehensive understanding of engineered tissue physiology.
Collapse
Affiliation(s)
- Venkat M Ramakrishnan
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hatim Thaker
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriel-Luis Ocampo
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rosalyn M Adam
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos R Estrada
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Roseti L, Cavallo C, Desando G, D’Alessandro M, Grigolo B. Forty Years of the Use of Cells for Cartilage Regeneration: The Research Side. Pharmaceutics 2024; 16:1622. [PMID: 39771600 PMCID: PMC11677864 DOI: 10.3390/pharmaceutics16121622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of articular cartilage damage has always represented a problem of considerable practical interest for orthopedics. Over the years, many surgical techniques have been proposed to induce the growth of repairing tissue and limit degeneration. In 1994, the turning point occurred: implanted autologous cells paved the way for a new treatment option based more on regeneration than repair. Objectives: This review aims to outline biological and clinical advances, from the use of mature adult chondrocytes to cell-derived products, going through progenitor cells derived from bone marrow or adipose tissue and their concentrates for articular cartilage repair. Moreover, it highlights the relevance of gene therapy as a valuable tool for successfully implementing current regenerative treatments, and overcoming the limitations of the local delivery of growth factors. Conclusions: Finally, this review concludes with an outlook on the importance of understanding the role and mechanisms of action of the different cell compounds with a view to implementing personalized treatments.
Collapse
Affiliation(s)
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.); (G.D.); (M.D.); (B.G.)
| | | | | | | |
Collapse
|
11
|
Li X, Zhang G, Wang M, Lu C, Zhang G, Chen Z, Ji Y. Comparison of stromal vascular fraction cell composition between Coleman fat and extracellular matrix/stromal vascular fraction gel. Adipocyte 2024; 13:2360037. [PMID: 38829527 PMCID: PMC11152101 DOI: 10.1080/21623945.2024.2360037] [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: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
As a mechanically condensed product of Coleman fat, extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel) eliminates adipocytes, concentrates SVF cells, and improves fat graft retention. This study aims to compare SVF cell composition between Coleman fat and ECM/SVF-gel. Matched Coleman fat and ECM/SVF-gel of 28 healthy women were subjected to RNA-seq, followed by functional enrichment and cell-type-specific enrichment analyses, and deconvolution of SVF cell subsets, reconstructing SVF cell composition in the transcriptome level. ECM/SVF-gels had 9 upregulated and 73 downregulated differentially expressed genes (DEGs). Downregulated DEGs were mainly associated with inflammatory and immune responses, and enriched in fat macrophages. M2 macrophages, resting CD4+ memory T cells, M1 macrophages, resting mast cells, and M0 macrophages ranked in the top five most prevalent immune cells in the two groups. The proportions of the principal non-immune cells (e.g., adipose-derived stem cells, pericytes, preadipocytes, microvascular endothelial cells) had no statistical differences between the two groups. Our findings reveal ECM/SVF-gels share the same dominant immune cells beneficial to fat graft survival with Coleman fat, but exhibiting obvious losses of immune cells (especially macrophages), while non-immune cells necessary for adipose regeneration might have no significant loss in ECM/SVF-gels and their biological effects could be markedly enhanced by the ECM/SVF-gel's condensed nature.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Mengmeng Wang
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Changhao Lu
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guangping Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Zhehui Chen
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yingchang Ji
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
12
|
Byun KA, Seo SB, Oh S, Jang JW, Son KH, Byun K. Poly-D,L-Lactic Acid Fillers Increase Subcutaneous Adipose Tissue Volume by Promoting Adipogenesis in Aged Animal Skin. Int J Mol Sci 2024; 25:12739. [PMID: 39684448 DOI: 10.3390/ijms252312739] [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: 11/10/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
During aging, subcutaneous white adipose tissue (sWAT) thickness and the adipogenic potential of adipose-derived stem cells (ASCs) decline. Poly-D,L-lactic acid (PDLLA) fillers are commonly used to restore diminished facial volume. Piezo1 increases polarizing macrophages towards the M2 phenotype, which promotes the secretion of fibroblast growth factor 2 (FGF2), thereby increasing ASC survival. This study evaluated whether PDLLA enhances adipogenesis in ASCs by modulating M2 polarization in an in vitro senescence model and in aged animals. Lipopolysaccharide (LPS)-induced senescent macrophages showed decreased Piezo1, which was upregulated by PDLLA. CD163 (an M2 marker) and FGF2 were downregulated in senescent macrophages but were upregulated by PDLLA. We evaluated whether reduced FGF2 secretion from senescent macrophages affects ASCs by applying conditioned media (CM) from macrophage cultures to ASCs. CM from senescent macrophages decreased ERK1/2 and proliferation in ASCs, both of which were restored by CM from PDLLA-stimulated senescent macrophages. Adipogenesis inducers (PPAR-γ and C/EBP-α) were downregulated by CM from senescent macrophages but upregulated by CM from PDLLA-stimulated senescent macrophages in ASCs. Similar patterns were observed in aged animal adipose tissue. PDLLA increased Piezo1 activity, M2 polarization, and FGF2 levels. PDLLA also enhanced ERK1/2, cell proliferation, PPAR-γ, and C/EBP-α expression, leading to increased adipose tissue thickness. In conclusion, our study showed that PDLLA increased adipose tissue thickness by modulating adipogenesis.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Suk Bae Seo
- SeoAh Song Dermatologic Clinic, Seoul 05557, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Won Jang
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
13
|
Ansari Lari S, Zumot MS, Nemrish S, Fredericks S. Role of mesenchymal cells in enhancing cosmetic outcomes for autologous augmented fat transfers for facial rejuvenation and reconstructive surgery. Front Med (Lausanne) 2024; 11:1466939. [PMID: 39610689 PMCID: PMC11602321 DOI: 10.3389/fmed.2024.1466939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
In recent years, autologous fat transfer (AFT) has gained popularity for reconstructive and cosmetic procedures due to its minimally invasive nature and natural-looking results. However, limitations such as unpredictable fat resorption and safety concerns persist. To address these issues, researchers have explored incorporating adipose-derived stromal cells (ADSCs) into fat grafts. Enriching fat grafts with ADSCs, often through stromal vascular fraction (SVF), shows promise in regenerative medicine, though their effectiveness remains debated. Some studies suggest no significant difference in outcomes, while others indicate that ADSCs are more effective in larger-volume grafts. This implies that ADSC-enriched grafts might achieve similar results to traditional methods, with volume retention being a crucial success indicator. Given that these cosmetic procedures impact body image and self-confidence, innovative techniques like ADSC-enriched grafts are crucial for improving clinical outcomes. ADSCs are favoured for their abundance in adipose tissue and wound healing properties, which enhance cosmetic results. Patients receiving ADSC-enriched grafts show increased collagen, elastin, and CD31 levels, and better graft survival compared to those with traditional fat grafting, reducing the need for repeat procedures. Recent applications in patients with fibrotic facial deformities have demonstrated positive outcomes both cosmetically and psychologically. This mini-review evaluates the efficacy and benefits of ADSC-enriched AFT for facial rejuvenation and reconstruction, focusing on graft retention and overall procedural outcomes.
Collapse
Affiliation(s)
- Soroush Ansari Lari
- Royal College of Surgeons in Ireland- Medical University of Bahrain, Muharraq, Bahrain
| | - Maya Salem Zumot
- Royal College of Surgeons in Ireland- Medical University of Bahrain, Muharraq, Bahrain
| | - Salsabiel Nemrish
- Royal College of Surgeons in Ireland- Medical University of Bahrain, Muharraq, Bahrain
| | - Salim Fredericks
- Department of Biochemistry, Royal College of Surgeons in Ireland- Medical University of Bahrain, Muharraq, Bahrain
| |
Collapse
|
14
|
Selestin Raja I, Kim C, Oh N, Park JH, Hong SW, Kang MS, Mao C, Han DW. Tailoring photobiomodulation to enhance tissue regeneration. Biomaterials 2024; 309:122623. [PMID: 38797121 DOI: 10.1016/j.biomaterials.2024.122623] [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: 02/07/2024] [Revised: 04/25/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Photobiomodulation (PBM), the use of biocompatible tissue-penetrating light to interact with intracellular chromophores to modulate the fates of cells and tissues, has emerged as a promising non-invasive approach to enhancing tissue regeneration. Unlike photodynamic or photothermal therapies that require the use of photothermal agents or photosensitizers, PBM treatment does not need external agents. With its non-harmful nature, PBM has demonstrated efficacy in enhancing molecular secretions and cellular functions relevant to tissue regeneration. The utilization of low-level light from various sources in PBM targets cytochrome c oxidase, leading to increased synthesis of adenosine triphosphate, induction of growth factor secretion, activation of signaling pathways, and promotion of direct or indirect gene expression. When integrated with stem cell populations, bioactive molecules or nanoparticles, or biomaterial scaffolds, PBM proves effective in significantly improving tissue regeneration. This review consolidates findings from in vitro, in vivo, and human clinical outcomes of both PBM alone and PBM-combined therapies in tissue regeneration applications. It encompasses the background of PBM invention, optimization of PBM parameters (such as wavelength, irradiation, and exposure time), and understanding of the mechanisms for PBM to enhance tissue regeneration. The comprehensive exploration concludes with insights into future directions and perspectives for the tissue regeneration applications of PBM.
Collapse
Affiliation(s)
| | - Chuntae Kim
- Institute of Nano-Bio Convergence, Pusan National University, Busan, 46241, Republic of Korea; Center for Biomaterials Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nuri Oh
- Department of Chemistry and Biology, Korea Science Academy of KAIST, Busan, 47162, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.
| | - Dong-Wook Han
- Institute of Nano-Bio Convergence, Pusan National University, Busan, 46241, Republic of Korea; Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
15
|
Xiao Y, Nie M, Xu W, Zhang J, Lei S, Wu D. The efficiency of human fat products in wound healing: A systematic review and meta-analysis. Int Wound J 2024; 21:e70016. [PMID: 39216014 PMCID: PMC11365526 DOI: 10.1111/iwj.70016] [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: 03/25/2024] [Revised: 07/14/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Wound development and healing involve intricate genetic and molecular processes, posing significant clinical management challenges. The objective of this study was to assess commonly used fat extracts' efficacy and safety (autologous fat, stromal vascular fraction and adipose-derived stem cells) in wound healing, particularly for refractory wounds, with the goal of providing evidence in clinical use. After a systematic review, 21 randomised controlled trials were included in our study. Based on the classification of human fat products, our meta-analysis revealed that the use of human fat products could speed healing rate, shorten healing time and achieve more complete healing, with statistically significant differences in outcome indicators when compared to conventional treatments. The analysis of histological findings across various studies indicated that fat extracts can promote epithelialization, collagen deposition and vascularization, thereby facilitating tissue regeneration and reducing inflammatory reactions. There were potential benefits to reducing patient pain levels after using adipose extracts. Furthermore, we analysed and summarised adverse events indicating the safe and effective clinical use of human fat products in wound treatment. Our research findings supported the efficiency of human fat products and demonstrated a high degree of safety in the clinical practice of wound management.
Collapse
Affiliation(s)
- Yutian Xiao
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| | - Mengqi Nie
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| | - Wenqing Xu
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| | - Jinglve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| | - Shaorong Lei
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| | - Dingyu Wu
- Department of Plastic and Cosmetic Surgery, Xiangya HospitalCentral South UniversityChangshaPR China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaPR China
| |
Collapse
|
16
|
Jeyaraman N, Shrivastava S, Ravi VR, Nallakumarasamy A, Pundkar A, Jeyaraman M. Understanding and controlling the variables for stromal vascular fraction therapy. World J Stem Cells 2024; 16:784-798. [PMID: 39219728 PMCID: PMC11362852 DOI: 10.4252/wjsc.v16.i8.784] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/13/2024] [Accepted: 07/25/2024] [Indexed: 08/26/2024] Open
Abstract
In regenerative medicine, the isolation of mesenchymal stromal cells (MSCs) from the adipose tissue's stromal vascular fraction (SVF) is a critical area of study. Our review meticulously examines the isolation process of MSCs, starting with the extraction of adipose tissue. The choice of liposuction technique, anatomical site, and immediate processing are essential to maintain cell functionality. We delve into the intricacies of enzymatic digestion, emphasizing the fine-tuning of enzyme concentrations to maximize cell yield while preventing harm. The review then outlines the filtration and centrifugation techniques necessary for isolating a purified SVF, alongside cell viability assessments like flow cytometry, which are vital for confirming the efficacy of the isolated MSCs. We discuss the advantages and drawbacks of using autologous vs allogeneic SVF sources, touching upon immunocompatibility and logistical considerations, as well as the variability inherent in donor-derived cells. Anesthesia choices, the selection between hypodermic needles vs liposuction cannulas, and the role of adipose tissue lysers in achieving cellular dissociation are evaluated for their impact on SVF isolation. Centrifugation protocols are also analyzed for their part in ensuring the integrity of the SVF. The necessity for standardized MSC isolation protocols is highlighted, promoting reproducibility and successful clinical application. We encourage ongoing research to deepen the understanding of MSC biology and therapeutic action, aiming to further the field of regenerative medicine. The review concludes with a call for rigorous research, interdisciplinary collaboration, and strict adherence to ethical and regulatory standards to safeguard patient safety and optimize treatment outcomes with MSCs.
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, Datta Meghe Institute of Higher Education and Research, Wardha 442004, Maharashtra, India
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India
| | - Sandeep Shrivastava
- Department of Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha 442004, Maharashtra, India
| | - V R Ravi
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, Datta Meghe Institute of Higher Education and Research, Wardha 442004, Maharashtra, India
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India
| | - Aditya Pundkar
- Department of Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha 442004, Maharashtra, India
| | - Madhan Jeyaraman
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
17
|
Sowa Y, Sawai S, Yamamoto K, Sunaga A, Saito N, Shirado T, Toyohara Y, Bolun L, Yoshimura K, Mazda O. Micronized cellular adipose matrix purified with a bladed connector contains abundant functional adipose stem cells. Tissue Cell 2024; 89:102457. [PMID: 38996772 DOI: 10.1016/j.tice.2024.102457] [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: 02/06/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024]
Abstract
INTRODUCTION A specialized device equipped with a sharp blade filter has been developed to enable more efficient purification of a micronized cellular adipose matrix (MCAM) containing stem cells. The aim of this study is to compare the characteristics and functions of the population of stromal cells (mSVF) and cultured cells (mASCs) purified using this device with those of cSVF and cASCs obtained through conventional enzymatic purification. METHODS Cell viability, proliferation capacity and yield were assessed. Characterization of stem cell potency was performed by analyzing cell surface markers including CD34, a marker of activated adipose-derived stem cells. The trilineage differentiation potential was evaluated using RT-PCR and histology. RESULTS The yield rate of mSVF obtained from MCAM was significantly higher than that with the conventional method, although use of the device resulted in a slight decrease in cell viability. After culture, mASCs exhibited a remarkable clonogenic potential and significantly higher cell proliferation potential than cASCs. The mASCs also displayed a distinct pattern of ASC cell surface markers, increased expression of genes related to CD34, high pluripotency, and a high trilineage differentiation ability. CONCLUSION The specialized device enhanced the yield of SVF and produced cells with high proliferation rates and characteristics that include expression of stem cell markers.
Collapse
Affiliation(s)
- Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Japan; Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Plastic and Reconstructive Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Seiji Sawai
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenta Yamamoto
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ataru Sunaga
- Department of Plastic Surgery, Jichi Medical University, Japan
| | - Natsumi Saito
- Department of Plastic Surgery, Jichi Medical University, Japan
| | - Takako Shirado
- Department of Plastic Surgery, Jichi Medical University, Japan
| | | | - Li Bolun
- Department of Plastic Surgery, Jichi Medical University, Japan
| | | | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Jacobs T, Mahoney C, Mohammed S, Ziccardi V. Evaluating Stromal Vascular Fraction As a Treatment for Peripheral Nerve Regeneration: A Scoping Review. J Oral Maxillofac Surg 2024; 82:771-781. [PMID: 38621666 DOI: 10.1016/j.joms.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE This study aims to investigate the potential of stromal vascular fraction (SVF) for peripheral nerve regeneration. METHODS A scoping review of Scopus and PubMed databases was conducted. Inclusion criteria were human or animal studies exploring the use of SVF for peripheral nerve regeneration. Studies were categorized by assessed outcomes: pain assessment, neural integrity, muscle recovery, and functional recovery. Level of evidence and study quality were assessed. RESULTS Nine studies met the inclusion criteria. SVF injection in humans with trigeminal neuropathic pain reduced pain scores from 7.5 ± 1.58 to 4.3 ± 3.28. SVF injection improved sensation in humans with leprosy neuropathy. Repairing transected rat sciatic nerves with SVF-coated nerve autografts improved wet muscle weight ratios (0.65 ± 0.11 vs 0.55 ± 0.06) and sciatic functional index (SFI) scores (-68.2 ± 9.2 vs -72.5 ± 8.9). Repairing transected rat sciatic nerves with SVF-coated conduits increased the ratio of gastrocnemius muscle weights (RGMW) (7-10% improvement), myelinated fibers (1,605 ± 806.2 vs 543.6 ± 478.66), and myelin thickness (5-20% increase). Repairing transected rat facial nerves with SVF-coated conduits improved whisker motion (9.22° ± 0.65° vs 1.90° ± 0.84°) and myelin thickness (0.57 μm ± 0.17 vs 0.45 μm ± 0.14 μm). Repairing transected rat sciatic nerves with SVF-coated nerve allografts improved RGMW (85 vs 50%), SFI scores (-20 to -10 vs -40 to -30), and Basso, Beatie, and Bresnahan locomotor scores (18 vs 15). All metrics mentioned above were statistically significant. The human studies were level 4 evidence due to being case series, while animal studies were the lowest level of evidence. CONCLUSION Despite initial promising results, the low-level evidence from the included studies warrants further investigation.
Collapse
Affiliation(s)
- Tyler Jacobs
- Resident, Department of Oral and Maxillofacial Surgery, Rutgers School of Dental Medicine, Newark, NJ.
| | | | - Saad Mohammed
- B.A. Candidate, New Jersey Institute of Technology, Newark, NJ
| | - Vincent Ziccardi
- Professor, Chair, and Associate Dean for Hospital Affairs, Department of Oral and Maxillofacial Surgery, Rutgers School of Dental Medicine, Newark, NJ
| |
Collapse
|
19
|
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
|
20
|
Yu W, Wang Z, Dai Y, Zhao S, Chen H, Wang S, Xie H. Autologous fat grafting for postoperative breast reconstruction: A systemic review. Regen Ther 2024; 26:1010-1017. [PMID: 39553540 PMCID: PMC11564784 DOI: 10.1016/j.reth.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Autologous fat grafting technology has become a new method for breast reconstruction after breast surgery due to its advantages of simple operation, low immunogenicity, fewer complications, high patient acceptance, and natural filling effect. However, the unpredictable fate of transplanted fat limits its widespread application. Currently, many studies have made certain progress in improving the survival rate of fat grafts. This article provides an overview of autologous fat grafting technology, including the mechanisms of fat graft survival, techniques for obtaining and transplanting adipose tissue, methods for enhancing graft survival, and complications associated with fat grafting.
Collapse
Affiliation(s)
| | | | - Yuhan Dai
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Shuhan Zhao
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Huilin Chen
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| |
Collapse
|
21
|
Gareev I, Beylerli O, Ilyasova T, Ahmad A, Shi H, Chekhonin V. Therapeutic application of adipose-derived stromal vascular fraction in myocardial infarction. iScience 2024; 27:109791. [PMID: 38736548 PMCID: PMC11088339 DOI: 10.1016/j.isci.2024.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024] Open
Abstract
The insufficiency of natural regeneration processes in higher organisms, including humans, underlies myocardial infarction (MI), which is one of the main causes of disability and mortality in the population of developed countries. The solution to this problem lies in the field of revealing the mechanisms of regeneration and creating on this basis new technologies for stimulating endogenous regenerative processes or replacing lost parts of tissues and organs with transplanted cells. Of great interest is the use of the so-called stromal vascular fraction (SVF), derived from autologous adipose tissue. It is known that the main functions of SVF are angiogenetic, antiapoptotic, antifibrotic, immune regulation, anti-inflammatory, and trophic. This study presents data on the possibility of using SVF, targeted regulation of its properties and reparative potential, as well as the results of research studies on its use for the restoration of damaged ischemic tissue after MI.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Bashkir State Medical University, Ufa 450008, Russian Federation
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa 450008, Russian Federation
| | - Tatiana Ilyasova
- Bashkir State Medical University, Ufa 450008, Russian Federation
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin 1500, China
| | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- The National Medical Research Center for Endocrinology, Moscow, Russian Federation
| |
Collapse
|
22
|
Kearns ML, Reynolds CM. Developmentally programmed obesity: Is there a role for anti-inflammatory nutritional strategies? Exp Physiol 2024; 109:633-646. [PMID: 38031876 PMCID: PMC11061634 DOI: 10.1113/ep091209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023]
Abstract
Pregnancy represents a period of immense maternal physiological adaptation, with progressive increases in lipid storage potential and insulin resistance to support fetal/placental growth. This requires significant change in the adipose tissue. Women living with obesity/overweight are more susceptible to these changes causing complications such as gestational diabetes. This is particularly worrying as up to 60% of European women are living with overweight/obesity at the onset of pregnancy. Furthermore, less than 1% meet all nutrition guidelines. There is now evidence that these deep metabolic changes can result in a predisposition to metabolic disease in both the mother and child in later life. Health and nutrition status during this period therefore represents a window to future health. This period offers a valuable opportunity for intervention to prevent the negative consequences of poor in utero environments and increases the long-term quality of life for mother and offspring. This review will examine a range of in utero factors which determine adipose tissue development, the impact of these factors on later-life obesity and metabolic health and the therapeutic value of dietary anti-inflammatory nutritional interventions during pregnancy and early life. When it comes to early life nutrition, a 'one size fits all' approach is not always appropriate. Understanding the mechanisms of adipose tissue development in response to differing nutritional strategies may be important in the context of complicated or adverse in utero environments and represents a substantial step towards a more personalised nutritional approach for the prevention of obesity, metabolic syndrome and related non-communicable diseases in future generations.
Collapse
Affiliation(s)
- Michelle L. Kearns
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| | - Clare M. Reynolds
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| |
Collapse
|
23
|
Josh F, Soekamto T, Windura C, Lumalessil D. The Combination of Stromal Vascular Fraction Cells and Platelet-Rich Plasma Increases Fibroblast Growth Factor 2 and Insulin-Like Growth Factor 1 in Full-Thickness Burns in Animal Model. ANNALS OF BURNS AND FIRE DISASTERS 2024; 37:35-44. [PMID: 38680831 PMCID: PMC11041884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/10/2023] [Indexed: 05/01/2024]
Abstract
The previous study on the injection of SVFs in combination with PRP showed positive effect on the healing of deep dermal burns. We now seek to understand the effect on full thickness burns, as assessed by changes in serum FGF2, IGF1, epithelialization, and fibroblast count. Forty-eight Wistar rats were randomly divided into four groups: (1) rats with full thickness burns given a local injection of combined SVFs and PRP; (2) rats with burns given topical Vaseline; (3) rats with burns given a local injection of placebo; and (4) rats without burns. Primary data were measured according to the time of euthanasia (at the 8th hour, 4th day, 7th day, 14th day or 21st day). One-way ANOVA test followed by post hoc test were used. Epithelialization in rats who received SVFs and PRP was superior on days 7, 14 and 21 when compared to the other groups. The fibroblast count in rats who received SVFs and PRP showed significant difference on days 7 (p=0.022). Significant differences in serum FGF2 were observed on days 4, 7, 14 and 21 (p=0.003, p=0.001, p=0.024, p=0.038, respectively). A significant difference was also observed in serum IGF1 levels on days 7, 14 and 21 (p=0.043, p=0.003, p=0.045, respectively), and the combination of SVFs and PRP showed superior results compared to other groups. Injection of combined SVFs and PRP increases FGF2, IGF1, fibroblast count, and epithelialization.
Collapse
Affiliation(s)
- F. Josh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - T.H. Soekamto
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Dr. Wahidin Sudirohusodo Hospital, Makassar, Indonesia
| | - C.A. Windura
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - D.G. Lumalessil
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
24
|
Mormone E, Cisternino A, Capone L, Caradonna E, Sbarbati A. The Model of Interstitial Cystitis for Evaluating New Molecular Strategies of Interstitial Regeneration in Humans. Int J Mol Sci 2024; 25:2326. [PMID: 38397003 PMCID: PMC10889234 DOI: 10.3390/ijms25042326] [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: 12/10/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Given the recent evidence in the clinical application of regenerative medicine, mostly on integumentary systems, we focused our interests on recent bladder regeneration approaches based on mesenchymal stem cells (MSCs), platelet-rich plasma (PRP), and hyaluronic acid (HA) in the treatment of interstitial cystitis/bladder pain syndrome (IC/BPS) in humans. IC/BPS is a heterogeneous chronic disease with not-well-understood etiology, characterized by suprapubic pain related to bladder filling and urothelium dysfunction, in which the impairment of immunological processes seems to play an important role. The histopathological features of IC include ulceration of the mucosa, edema, denuded urothelium, and increased detection of mast cells and other inflammatory cells. A deeper understanding of the molecular mechanism underlying this disease is essential for the selection of the right therapeutic approach. In fact, although various therapeutic strategies exist, no efficient therapy for IC/BPS has been discovered yet. This review gives an overview of the clinical and pathological features of IC/BPS, with a particular focus on the molecular pathways involved and a special interest in the ongoing few investigational therapies in IC/BPS, which use new regenerative medicine approaches, and their synergetic combination. Good knowledge of the molecular aspects related to stem cell-, PRP-, and biomaterial-based treatments, as well as the understanding of the molecular mechanism of this pathology, will allow for the selection of the right and best use of regenerative approaches of structures involving connective tissue and epithelia, as well as in other diseases.
Collapse
Affiliation(s)
- Elisabetta Mormone
- Intitute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Italy
| | - Antonio Cisternino
- Santa Maria di Bari Hospital, Via Antonio de Ferraris 22, 70124 Bari, Italy;
| | - Lorenzo Capone
- Department of Urology, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini 1, 71013 San Giovanni Rotondo, Italy;
| | | | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37129 Verona, Italy;
| |
Collapse
|
25
|
Ertlen C, Seblani M, Bonnet M, Brezun JM, Coyle T, Sabatier F, Fuentes S, Decherchi P, Serratrice N, Marqueste T. Efficacy of the immediate adipose-derived stromal vascular fraction autograft on functional sensorimotor recovery after spinal cord contusion in rats. Stem Cell Res Ther 2024; 15:29. [PMID: 38303017 PMCID: PMC10835949 DOI: 10.1186/s13287-024-03645-z] [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: 09/13/2023] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Spinal cord injuries (SCI) lead to functional alteration with important consequences such as motor and sensory disorders. The repair strategies developed to date remain ineffective. The adipose tissue-derived stromal vascular fraction (SVF) is composed of a cocktail of cells with trophic, pro-angiogenic and immunomodulatory effects. Numerous therapeutic benefits were shown for tissue reconstitution, peripheral neuropathy and for the improvement of neurodegenerative diseases. Here, the therapeutic efficacy of SVF on sensorimotor recovery after an acute thoracic spinal cord contusion in adult rats was determined. METHOD Male Sprague Dawley rats (n = 45) were divided into 3 groups: SHAM (without SCI and treatment), NaCl (animals with a spinal lesion and receiving a saline injection through the dura mater) and SVF (animals with a spinal lesion and receiving a fraction of fat removed from adipocytes through the dura mater). Some animals were sacrificed 14 days after the start of the experiment to determine the inflammatory reaction by measuring the interleukin-1β, interleukin-6 and Tumor Necrosis Factor-α in the lesion area. Other animals were followed once a week for 12 weeks to assess functional recovery (postural and locomotor activities, sensorimotor coordination). At the end of this period, spinal reflexivity (rate-dependent depression of the H-reflex) and physiological adjustments (ventilatory response to metabosensitive muscle activation following muscle fatigue) were measured with electrophysiological tools. RESULTS Compared to non-treated animals, results indicated that the SVF reduced the endogenous inflammation and increased the behavioral recovery in treated animals. Moreover, H-reflex depression and ventilatory adjustments to muscle fatigue were found to be comparable between SHAM and SVF groups. CONCLUSION Our results highlight the effectiveness of SVF and its high therapeutic potential to improve sensorimotor functions and to restore the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions after traumatic contusion.
Collapse
Affiliation(s)
- 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Florence Sabatier
- Assistance Publique - Hôpitaux de Marseille (AP-HM), INSERM 1409 Centre d'Investigation Clinique en Biothérapies, Unité de Culture Et Thérapie Cellulaire, Hôpital de La Conception, 147, Boulevard Baille, 13385, Marseille Cedex 05, France
| | - Stéphane Fuentes
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| | - Nicolas Serratrice
- 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, Marseille, France
| | - 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, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| |
Collapse
|
26
|
Neubauer M, Otahal A, Kuten O, Sherman SL, Moser L, Kramer K, DeLuna A, Neugebauer J, Dammerer D, Muellner T, Nehrer S. Infra-patellar fat pad-derived mesenchymal stem cells maintain their chondrogenic differentiation potential after arthroscopic harvest with blood-product supplementation. INTERNATIONAL ORTHOPAEDICS 2024; 48:279-290. [PMID: 37646823 PMCID: PMC10766657 DOI: 10.1007/s00264-023-05930-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/06/2023] [Indexed: 09/01/2023]
Abstract
PURPOSE Mesenchymal stem cells/medicinal signaling cells (MSCs) possess therapeutic potential and are used in regenerative orthopaedics. The infra-patellar fat pad (IFP) is partially resected during knee arthroscopy (KASC) and contains MSCs. Heat, irrigation, and mechanical stress during KASC may decrease MSC's therapeutic potential. This study assessed MSCs' regenerative potential after arthroscopic IFP harvest and potential effects of two blood products (BP) (platelet-rich plasma (PRP), hyperacute serum (HAS)) on MSCs' viability and chondrogenic differentiation capacity. METHODS IFP was arthroscopically harvested, isolated, and counted (n = 5). Flow cytometry was used to assess cell viability via staining with annexin V/7-AAD and stemness markers via staining for CD90, CD73, and CD105. MSCs were incubated with blood products, and metabolic activity was determined via an XTT assay. Deposition of cartilage extracellular matrix was determined in histologic sections of chondrogenically differentiated 3D pellet cultures via staining with Alcian Blue. Expression of cartilage-specific genes (SOX9, MMP3/13, ACAN, COL1/2) was analyzed via quantitative PCR. RESULTS MSC isolation from IFP yielded 2.66*106 ± 1.49*106 viable cells from 2.7 (0.748) g of tissue. MSC markers (CD 90/105/73) were successfully detected and annexin V staining showed 81.5% viable cells. XTT showed increased metabolic activity. Within the BP groups, this increase was significant (days 0-14, p < 0.05). PCR showed expression of cartilage-specific genes in each group. COL2 (p < 0.01) as well as ACAN (p < 0.001) expression levels were significantly higher in the HAS group. Histology showed successful differentiation. CONCLUSION Arthroscopic harvest of IFP-MSCs yields sufficient cells with maintained regenerative potential and viability. Blood products further enhance MSCs' viability.
Collapse
Affiliation(s)
- Markus Neubauer
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
- Division of Orthopaedics and Traumatology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500, Krems, Austria
| | - Alexander Otahal
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
| | - Olga Kuten
- Ortho Sera GmbH, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
| | | | - Lukas Moser
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
- Division of Orthopaedics and Traumatology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500, Krems, Austria
| | - Karina Kramer
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
| | - Andrea DeLuna
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
| | - Johannes Neugebauer
- Division of Orthopaedics and Traumatology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500, Krems, Austria
| | - Dietmar Dammerer
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
- Division of Orthopaedics and Traumatology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500, Krems, Austria
| | - Thomas Muellner
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria
- Department of Orthopaedics and Traumatology, Evangelic Hospital Vienna, Hans-Sachs-Gasse 10-12, 1180, Vienna, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine and Orthopaedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Austria.
- Division of Orthopaedics and Traumatology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500, Krems, Austria.
| |
Collapse
|
27
|
Tantuway V, Jeyaraman M, Nallakumarasamy A, Prikh MB, Sharma AK, Sharma R. Functional Outcome Analysis of Autologous Stromal Vascular Fraction (SVF) (Sahaj Therapy ®) Using Direct Sonication in Osteonecrosis of the Femoral Head (ONFH): A 6-Year Follow-Up Study. Indian J Orthop 2024; 58:68-78. [PMID: 38161400 PMCID: PMC10754810 DOI: 10.1007/s43465-023-01041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/28/2023] [Indexed: 01/03/2024]
Abstract
Introduction We investigated the safety, efficacy, functional, and clinical outcomes of intra-osseous implantation of mechanically isolated, autologous stromal vascular fraction (SVF), an Australian patented direct ultrasonication technology (Sahaj Therapy®) in osteonecrosis of the femoral head (ONFH). Materials and Methods A total of 32 cases of ONFH were enrolled in the study after confirming with an MRI of the affected hip. All cases were treated with an intra-osseous autologous SVF implantation [4-5 cc with the cellular dosage of 8.0 × 107 cells with a viability of > 85% SVF cells] on the same surgical sitting. All the cases were followed up clinically, functionally, and radiologically at regular intervals. A comparison of mean HOOS scores at different follow-ups was done using Paired 't'-test. A P value of < 0.05 was considered significant. Results In our study, male preponderance was seen (53.1%). According to the modified Ficat and Arlet classification, the most common grade of ONFH was grade 2 [right: 25 hips and left: 25 hips]. There was a statistically significant improvement in the mean HOOS score of the right hip (n = 10) and left hip (n = 9) from preoperative time till 72 months (P < 0.05). The follow-up MRI of the affected hips shows improved osteogenesis without any further worsening of the contour of the femoral head. No adverse effects were seen in any of the study participants. Conclusion For individuals with ONFH, treated with intra-osseous autologous SVF implantation in the same surgical procedure is an innovative and promising treatment modality. Even after 6 years of follow-up, the study participants with ONFH have shown good clinical and functional outcomes with autologous SVF.
Collapse
Affiliation(s)
- Vinay Tantuway
- Department of Orthopaedics and Traumatology, Index Medical College Hospital and Research Centre, Indore, Madhya Pradesh India
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| | - Madhan Jeyaraman
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX USA
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
| | - Mittal B. Prikh
- Department of Orthopaedics, Navjivan Hospital, Ahmedabad, Gujarat India
| | - Aashish K. Sharma
- Department of Orthopaedics and Joint Replacement, CK Birla Hospitals, Jaipur, Rajasthan India
| | - Raj Sharma
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| |
Collapse
|
28
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Kesharwani D, Brown AC. Navigating the Adipocyte Precursor Niche: Cell-Cell Interactions, Regulatory Mechanisms and Implications for Adipose Tissue Homeostasis. JOURNAL OF CELLULAR SIGNALING 2024; 5:65-86. [PMID: 38826152 PMCID: PMC11141760 DOI: 10.33696/signaling.5.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Support for stem cell self-renewal and differentiation hinges upon the intricate microenvironment termed the stem cell 'niche'. Within the adipose tissue stem cell niche, diverse cell types, such as endothelial cells, immune cells, mural cells, and adipocytes, intricately regulate the function of adipocyte precursors. These interactions, whether direct or indirect, play a pivotal role in governing the balance between self-renewal and differentiation of adipocyte precursors into adipocytes. The mechanisms orchestrating the maintenance and coordination of this niche are still in the early stages of comprehension, despite their crucial role in regulating adipose tissue homeostasis. The complexity of understanding adipocyte precursor renewal and differentiation is amplified due to the challenges posed by the absence of suitable surface receptors for identification, limitations in creating optimal ex vivo culture conditions for expansion and constraints in conducting in vivo studies. This review delves into the current landscape of knowledge surrounding adipocyte precursors within the adipose stem cell niche. We will review the identification of adipocyte precursors, the cell-cell interactions they engage in, the factors influencing their renewal and commitment toward adipocytes and the transformations they undergo during instances of obesity.
Collapse
Affiliation(s)
- Devesh Kesharwani
- Center for Molecular Medicine, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Aaron C. Brown
- Center for Molecular Medicine, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
- School of Biomedical Sciences and Engineering, The University of Maine, Orono, Maine 04469, USA
- Tufts University School of Medicine, 145 Harrison Ave, Boston, MA 02111, USA
| |
Collapse
|
30
|
Bi M, Yang K, Yu T, Wu G, Li Q. Cell-based mechanisms and strategies of co-culture system both in vivo and vitro for bone tissue engineering. Biomed Pharmacother 2023; 169:115907. [PMID: 37984308 DOI: 10.1016/j.biopha.2023.115907] [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: 09/09/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The lack of a functional vascular supply has been identified as a major challenge limiting the clinical introduction of stem cell-based bone tissue engineering (BTE) for the repair of large-volume bone defects (LVBD). Various approaches have been explored to improve the vascular supply in tissue-engineered constructs, and the development of strategies that could effectively induce the establishment of a functional vascular supply has become a major goal of BTE research. One of the state-of-the-art methods is to incorporate both angiogenic and osteogenic cells in co-culture systems. This review clarifies the key concepts involved, summarises the cell types and models used to date, and systematically evaluates their performance. We also discuss the cell-to-cell communication between these two cell types and the strategies explored in BTE constructs with angiogenic and osteogenic cells to optimise their functions. In addition, we outline unresolved issues and remaining obstacles that need to be overcome for further development in this field and eventual successful repair of LVBD.
Collapse
Affiliation(s)
- Mengning Bi
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology Shanghai, China
| | - Kaiwen Yang
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Key Laboratory of Stomatology &Shanghai Research Institute of Stomatology; National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, the Netherlands.
| | - Qiong Li
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
31
|
Villanueva-Carmona T, Cedó L, Núñez-Roa C, Maymó-Masip E, Vendrell J, Fernández-Veledo S. Protocol for the in vitro isolation and culture of mature adipocytes and white adipose tissue explants from humans and mice. STAR Protoc 2023; 4:102693. [PMID: 37924518 PMCID: PMC10656257 DOI: 10.1016/j.xpro.2023.102693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/10/2023] [Accepted: 10/12/2023] [Indexed: 11/06/2023] Open
Abstract
White adipose tissue (WAT) explants culture allows the study of this tissue ex vivo, maintaining its structure and properties. Concurrently, isolating mature adipocytes facilitates research into fat cell metabolism and hormonal regulation. Here, we present a protocol for obtaining, isolating, and processing mature adipocytes, alongside the cultivation of WAT explants from humans and mice. We describe steps for WAT retrieval, culturing of WAT explants, WAT digestion, and adipocytes separation. We then detail procedures for culturing isolated mature adipocytes. For complete details on the use and execution of this protocol, please refer to Villanueva-Carmona et al. (2023).1.
Collapse
Affiliation(s)
- Teresa Villanueva-Carmona
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), 43201 Reus, Spain
| | - Lídia Cedó
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Catalina Núñez-Roa
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elsa Maymó-Masip
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Joan Vendrell
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), 43201 Reus, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
32
|
Pirraco RP. Macro, Micro, and Everything in Between. Bridging the Gap: A Vision Toward the Creation of Multiscale Vascular Networks. Adv Biol (Weinh) 2023; 7:e2300291. [PMID: 37566782 DOI: 10.1002/adbi.202300291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/27/2023] [Indexed: 08/13/2023]
Abstract
Vascularization is a key issue for the clinical translation of tissue engineering strategies. This has been recognized in the field for almost two decades. Several strategies to solve this issue are proposed but none has decisively tackled the problem. This is in part due to an excessive focus on microvascularization that ignores the need of having macrovessels capable of being surgically connected to the patient's circulation upon implantation. Indeed, a strategy for macrovessel engineering must co-exist with a strategy for microvessels. And if this is true, all the intermediate scales have to be addressed as well. Therefore, multiscale vascular networks must be the focus of tissue engineering vascularization efforts. In this work, a reflection is made on a possible path forward for researchers and engineers in the field to achieve the ultimate goal of efficient vascularization of engineered tissues and organs.
Collapse
Affiliation(s)
- Rogério Pedro Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, 4805-017, Portugal
| |
Collapse
|
33
|
Escudero M, Vaysse L, Eke G, Peyrou M, Villarroya F, Bonnel S, Jeanson Y, Boyer L, Vieu C, Chaput B, Yao X, Deschaseaux F, Parny M, Raymond‐Letron I, Dani C, Carrière A, Malaquin L, Casteilla L. Scalable Generation of Pre-Vascularized and Functional Human Beige Adipose Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301499. [PMID: 37731092 PMCID: PMC10625054 DOI: 10.1002/advs.202301499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/07/2023] [Indexed: 09/22/2023]
Abstract
Obesity and type 2 diabetes are becoming a global sociobiomedical burden. Beige adipocytes are emerging as key inducible actors and putative relevant therapeutic targets for improving metabolic health. However, in vitro models of human beige adipose tissue are currently lacking and hinder research into this cell type and biotherapy development. Unlike traditional bottom-up engineering approaches that aim to generate building blocks, here a scalable system is proposed to generate pre-vascularized and functional human beige adipose tissue organoids using the human stromal vascular fraction of white adipose tissue as a source of adipose and endothelial progenitors. This engineered method uses a defined biomechanical and chemical environment using tumor growth factor β (TGFβ) pathway inhibition and specific gelatin methacryloyl (GelMA) embedding parameters to promote the self-organization of spheroids in GelMA hydrogel, facilitating beige adipogenesis and vascularization. The resulting vascularized organoids display key features of native beige adipose tissue including inducible Uncoupling Protein-1 (UCP1) expression, increased uncoupled mitochondrial respiration, and batokines secretion. The controlled assembly of spheroids allows to translate organoid morphogenesis to a macroscopic scale, generating vascularized centimeter-scale beige adipose micro-tissues. This approach represents a significant advancement in developing in vitro human beige adipose tissue models and facilitates broad applications ranging from basic research to biotherapies.
Collapse
Affiliation(s)
- Mélanie Escudero
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Laurence Vaysse
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Gozde Eke
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Marion Peyrou
- CIBER “Fisiopatologia de la Obesidad y Nutrición”, Department of Biochemistry and Molecular BiomedicineUniversity of BarcelonaMadrid28029Spain
| | - Francesc Villarroya
- CIBER “Fisiopatologia de la Obesidad y Nutrición”, Department of Biochemistry and Molecular BiomedicineUniversity of BarcelonaMadrid28029Spain
| | - Sophie Bonnel
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Yannick Jeanson
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Louisa Boyer
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Christophe Vieu
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Benoit Chaput
- Service de Chirurgie plastique, reconstructrice et esthétiqueCentre Hospitalier Universitaire RangueilToulouse31400France
| | - Xi Yao
- Faculté de MédecineUniversité Côte d'AzurINSERM, CNRS, iBVNice06103France
| | - Frédéric Deschaseaux
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Mélissa Parny
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LabHPEC, Histology and Pathology DepartmentUniversité de Toulouse, ENVTToulouse31076France
| | - Isabelle Raymond‐Letron
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LabHPEC, Histology and Pathology DepartmentUniversité de Toulouse, ENVTToulouse31076France
| | - Christian Dani
- Faculté de MédecineUniversité Côte d'AzurINSERM, CNRS, iBVNice06103France
| | - Audrey Carrière
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | | | - Louis Casteilla
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| |
Collapse
|
34
|
Tantuway V, Thomas W, Parikh MB, Sharma R, Jeyaraman N, Jeyaraman M. Clinical Outcome of Minimally Manipulated, Mechanically Isolated Autologous Adipose Tissue-Derived Stromal Vascular Fraction (Sahaj Therapy®) in Knee Osteoarthritis-Randomized Controlled Trial. Indian J Orthop 2023; 57:1646-1658. [PMID: 37766954 PMCID: PMC10519910 DOI: 10.1007/s43465-023-00981-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Introduction Regenerative therapy has shown promising results in the treatment of osteoarthritis (OA) knee with Kellgren-Lawrence (KL) Grades I-III. We compared the safety, efficacy, functional, and clinical outcomes of intra-articular implantation of autologous adipose tissue-derived stromal vascular fraction (SVF) isolated using direct ultrasonic cavitation (Sahaj therapy-Cell Innovation Patented Technology) and saline injection in knee osteoarthritis. Materials and Methods The present prospective observational study was conducted over 3 years. We enrolled 120 patients in our study, where four patients got excluded as they did not meet the inclusion criteria. The remaining 116 patients were randomized into two groups, one with autologous adipose tissue-derived SVF and the other group with saline injection. A comparison of mean KOOS and VAS scores at different follow-ups was done using Paired 't' test. A p value of < 0.05 was considered significant. Results The results show that the SVF group had significantly higher KOOS scores (78.49 ± 6.54 in the SVF group vs 59.19 ± 5.14 in the saline group), respectively (p < 0.001). Similarly, the SVF group had significantly lesser VAS scores (3.17 ± 0.94 in the SVF group vs 3.89 ± 1.04 in the saline group), respectively (p < 0.001). Conclusions Autologous adipose tissue-derived SVF is a better choice for treating knee osteoarthritis. For individuals with degenerative osteoarthritis, autologous SVF grafting in the same surgical procedure is an innovative and promising treatment modality. Even after 3 years of follow-up, the study participants with OA knee have shown a good clinical and functional outcome.
Collapse
Affiliation(s)
- Vinay Tantuway
- Department of Orthopaedics & Traumatology, Index Medical College Hospital and Research Centre, Indore, Madhya Pradesh India
| | - Wayne Thomas
- Department of Tumor Immunology, The University of Newcastle, Callaghan, Australia
| | - Mittal B. Parikh
- Department of Orthopaedics, Navjivan Hospital, Ahmedabad, Gujarat India
| | - Raj Sharma
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
| |
Collapse
|
35
|
Malekzadeh H, Tirmizi Z, Arellano JA, Egro FM, Ejaz A. Application of Adipose-Tissue Derived Products for Burn Wound Healing. Pharmaceuticals (Basel) 2023; 16:1302. [PMID: 37765109 PMCID: PMC10534650 DOI: 10.3390/ph16091302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Burn injuries are a significant global health concern, leading to high morbidity and mortality. Deep burn injuries often result in delayed healing and scar formation, necessitating effective treatment options. Regenerative medicine, particularly cell therapy using adipose-derived stem cells (ASCs), has emerged as a promising approach to improving burn wound healing and reducing scarring. Both in vitro and preclinical studies have demonstrated the efficacy of ASCs and the stromal vascular fraction (SVF) in addressing burn wounds. The application of ASCs for burn healing has been studied in various forms, including autologous or allogeneic cells delivered in suspension or within scaffolds in animal burn models. Additionally, ASC-derived non-cellular components, such as conditioned media or exosomes have shown promise. Injection of ASCs and SVF at burn sites have been demonstrated to enhance wound healing by reducing inflammation and promoting angiogenesis, epithelialization, and granulation tissue formation through their paracrine secretome. This review discusses the applications of adipose tissue derivatives in burn injury treatment, encompassing ASC transplantation, as well as the utilization of non-cellular components utilization for therapeutic benefits. The application of ASCs in burn healing in the future will require addressing donor variability, safety, and efficacy for successful clinical application.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
36
|
Vasella M, Arnke K, Dranseikiene D, Guzzi E, Melega F, Reid G, Klein HJ, Schweizer R, Tibbitt MW, Kim BS. Methacrylated Gelatin as a Scaffold for Mechanically Isolated Stromal Vascular Fraction for Cutaneous Wound Repair. Int J Mol Sci 2023; 24:13944. [PMID: 37762247 PMCID: PMC10530931 DOI: 10.3390/ijms241813944] [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: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a highly interesting cell source for regenerative purposes, including wound healing, and a practical alternative to enzymatically isolated SVF. In the clinical context, SVF benefits from scaffolds that facilitate viability and other cellular properties. In the present work, the feasibility of methacrylated gelatin (GelMA), a stiffness-tunable, light-inducible hydrogel with high biocompatibility is investigated as a scaffold for SVF in an in vitro setting. Lipoaspirates from elective surgical procedures were collected and processed to mSVF and mixed with GelMA precursor solutions. Non-encapsulated mSVF served as a control. Viability was measured over 21 days. Secreted basic fibroblast growth factor (bFGF) levels were measured on days 1, 7 and 21 by ELISA. IHC was performed to detect VEGF-A, perilipin-2, and CD73 expression on days 7 and 21. The impact of GelMA-mSVF on human dermal fibroblasts was measured in a co-culture assay by the same viability assay. The viability of cultured GelMA-mSVF was significantly higher after 21 days (p < 0.01) when compared to mSVF alone. Also, GelMA-mSVF secreted stable levels of bFGF over 21 days. While VEGF-A was primarily expressed on day 21, perilipin-2 and CD73-positive cells were observed on days 7 and 21. Finally, GelMA-mSVF significantly improved fibroblast viability as compared with GelMA alone (p < 0.01). GelMA may be a promising scaffold for mSVF as it maintains cell viability and proliferation with the release of growth factors while facilitating adipogenic differentiation, stromal cell marker expression and fibroblast proliferation.
Collapse
Affiliation(s)
- Mauro Vasella
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Kevin Arnke
- Center for Preclinical Development, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Dalia Dranseikiene
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Elia Guzzi
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Francesca Melega
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Gregory Reid
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Holger Jan Klein
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland;
| | - Riccardo Schweizer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Regional Hospital Lugano, 6900 Lugano, Switzerland;
| | - Mark W. Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| |
Collapse
|
37
|
Lee DK, Kim M, Jeong J, Lee YS, Yoon JW, An MJ, Jung HY, Kim CH, Ahn Y, Choi KH, Jo C, Lee CK. Unlocking the potential of stem cells: Their crucial role in the production of cultivated meat. Curr Res Food Sci 2023; 7:100551. [PMID: 37575132 PMCID: PMC10412782 DOI: 10.1016/j.crfs.2023.100551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Cellular agriculture is an emerging research field of agribiotechnology that aims to produce agricultural products using stem cells, without sacrificing animals or cultivating crops. Cultivated meat, as a representative cellular product of cellular agriculture, is being actively researched due to global food insecurity, environmental, and ethical concerns. This review focuses on the application of stem cells, which are the seeds of cellular agriculture, for the production of cultivated meat, with emphasis on deriving and culturing muscle and adipose stem cells for imitating fresh meat. Establishing standards and safety regulations for culturing stem cells is crucial for the market entry of cultured muscle tissue-based biomaterials. Understanding stem cells is a prerequisite for creating reliable cultivated meat and other cellular agricultural biomaterials. The techniques and regulations from the cultivated meat industry could pave the way for new cellular agriculture industries in the future.
Collapse
Affiliation(s)
- Dong-Kyung Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Minsu Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young-Seok Lee
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Ji Won Yoon
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Min-Jeong An
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Hyun Young Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cho Hyun Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yelim Ahn
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwang-Hwan Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| |
Collapse
|
38
|
Noël D, Scherberich A. Editorial: Biology and clinical applications of adipose-derived cells for skeletal regeneration. Front Bioeng Biotechnol 2023; 11:1221444. [PMID: 37288354 PMCID: PMC10242165 DOI: 10.3389/fbioe.2023.1221444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Affiliation(s)
- Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Arnaud Scherberich
- Bone Regeneration, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| |
Collapse
|
39
|
Ni X, Xing X, Deng Y, Li Z. Applications of Stimuli-Responsive Hydrogels in Bone and Cartilage Regeneration. Pharmaceutics 2023; 15:pharmaceutics15030982. [PMID: 36986842 PMCID: PMC10056098 DOI: 10.3390/pharmaceutics15030982] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Bone and cartilage regeneration is an area of tremendous interest and need in health care. Tissue engineering is a potential strategy for repairing and regenerating bone and cartilage defects. Hydrogels are among the most attractive biomaterials in bone and cartilage tissue engineering, mainly due to their moderate biocompatibility, hydrophilicity, and 3D network structure. Stimuli-responsive hydrogels have been a hot topic in recent decades. They can respond to external or internal stimulation and are used in the controlled delivery of drugs and tissue engineering. This review summarizes current progress in the use of stimuli-responsive hydrogels in bone and cartilage regeneration. The challenges, disadvantages, and future applications of stimuli-responsive hydrogels are briefly described.
Collapse
Affiliation(s)
- Xiaoqi Ni
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xin Xing
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yunfan Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhi Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
40
|
Hofmann E, Schwarz A, Fink J, Kamolz LP, Kotzbeck P. Modelling the Complexity of Human Skin In Vitro. Biomedicines 2023; 11:biomedicines11030794. [PMID: 36979772 PMCID: PMC10045055 DOI: 10.3390/biomedicines11030794] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023] Open
Abstract
The skin serves as an important barrier protecting the body from physical, chemical and pathogenic hazards as well as regulating the bi-directional transport of water, ions and nutrients. In order to improve the knowledge on skin structure and function as well as on skin diseases, animal experiments are often employed, but anatomical as well as physiological interspecies differences may result in poor translatability of animal-based data to the clinical situation. In vitro models, such as human reconstructed epidermis or full skin equivalents, are valuable alternatives to animal experiments. Enormous advances have been achieved in establishing skin models of increasing complexity in the past. In this review, human skin structures are described as well as the fast evolving technologies developed to reconstruct the complexity of human skin structures in vitro.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
41
|
Morito S, Kawasaki M, Nishiyama M, Sakumoto T, Hashiguchi M, Narita T, Kawaguchi A, Toda S, Aoki S. Microenvironmental elements singularity synergistically regulate the behavior and chemosensitivity of endometrioid carcinoma. Hum Cell 2023; 36:1147-1159. [PMID: 36853404 DOI: 10.1007/s13577-023-00886-7] [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: 12/27/2022] [Accepted: 02/23/2023] [Indexed: 03/01/2023]
Abstract
The importance of the microenvironment is widely recognized as it regulates not only malignant cell behavior but also drug sensitivity. The cancer cell microenvironment is composed of biological, physical and chemical elements, and simultaneous reproduction of these three elements are important conditions investigated in cancer research. In the present study, we focused on the epidemiological and anatomical specificities of endometrioid carcinoma, obesity (biological), fluid flow (physical) and anticancer agents (chemical) to target the specific microenvironmental elements of endometrioid carcinoma. To elucidate the individual effects of these elements on endometrioid carcinoma and to investigate the relationships between these factors, we developed an adipose tissue fragments (ATFs)-embedded cell disc under a rotational culture method to generate carcinoma-stroma interactions and to create fluid flow. ATFs and fluid flow individually or synergistically influenced proliferative cellular behavior and the morphological changes underlying endometrioid carcinoma. ATFs and fluid flow also governed the expression of extracellular signal-regulated kinase and p38 signaling synergistically or individually, depending on the endometrioid carcinoma cell type. Adipose tissue induced chemoresistance to cis-diamminedichloro-platinum (CDDP) in endometrioid cancer, but the resistance effect was abolished by fluid flow. Thus, a simple reconstructed model was established to investigate three elements of the microenvironment of endometrioid carcinoma in vitro. This culture model unequivocally demonstrated the individual and synergistic effects of the three elements on endometrioid carcinoma. This new culture model is a promising tool for elucidating the mechanisms underlying endometrioid carcinoma and for developing further treatment strategies.
Collapse
Affiliation(s)
- Sayuri Morito
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Maki Kawasaki
- Department of Urology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Megumi Nishiyama
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Takehisa Sakumoto
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Mariko Hashiguchi
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Takayuki Narita
- Department of Chemistry and Applied Chemistry, Faculty of Science and Engineering, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Atsushi Kawaguchi
- Education and Research Center for Community Medicine, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Shuji Toda
- Department of Pathology, Takagi Hospital, Okawa, Fukuoka, 831-8501, Japan
| | - Shigehisa Aoki
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan.
| |
Collapse
|
42
|
Musa M, Zeppieri M, Enaholo ES, Salati C, Parodi PC. Adipose Stem Cells in Modern-Day Ophthalmology. Clin Pract 2023; 13:230-245. [PMID: 36826163 PMCID: PMC9955457 DOI: 10.3390/clinpract13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Stem cells (SCs) have evolved as an interesting and viable factor in ophthalmologic patient care in the past decades. SCs have been classified as either embryonic, mesenchymal, tissue-specific, or induced pluripotent cells. Multiple novel management techniques and clinical trials have been established to date. While available publications are predominantly animal-model-based, significant material is derived from human studies and case-selected scenarios. This possibility of explanting cells from viable tissue to regenerate/repair damaged tissue points to an exciting future of therapeutic options in all fields of medicine, and ophthalmology is surely not left out. Adipose tissue obtained from lipo-aspirates has been shown to produce mesenchymal SCs that are potentially useful in different body parts, including the oculo-visual system. An overview of the anatomy, physiology, and extraction process for adipose-tissue-derived stem cells (ADSC) is important for better understanding the potential therapeutic benefits. This review examines published data on ADSCs in immune-modulatory, therapeutic, and regenerative treatments. We also look at the future of ADSC applications for ophthalmic patient care. The adverse effects of this relatively novel therapy are also discussed.
Collapse
Affiliation(s)
- Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Nigeria
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | | | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
43
|
Rudyk M, Hurmach Y, Serhiichuk T, Akulenko I, Skivka L, Berehova T, Ostapchenko L. Multi-probiotic consumption sex-dependently interferes with MSG-induced obesity and concomitant phagocyte pro-inflammatory polarization in rats: Food for thought about personalized nutrition. Heliyon 2023; 9:e13381. [PMID: 36816299 PMCID: PMC9932736 DOI: 10.1016/j.heliyon.2023.e13381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Epidemic scope which obesity has reached in many countries necessitates shifting the emphasis in medicine from traditional reaction to individualized and personalized prevention. Numerous trials convincingly prove sexual dimorphism of obesity in morbidity, pathophysiology, comorbidity, outcomes and prophylaxis efficacy. Obesity is characterized by chronic systemic low-grade inflammation that creates the preconditions for the emergence of numerous comorbidities. Leading role in the initiation, propagation and resolution of inflammation belongs to tissue resident and circulating phagocytes. The outcome of inflammation largely depends on phagocyte functional polarization, which in turn is governed by environmental stimuli. Gut microbiota (GM), whose disturbances are one of the key pathogenetic features in obesity, substantially affect phagocyte functions and can either aggravate or calm obesity-associated inflammation. Probiotics possess promising physiological functions, including microbiota-restoring and anti-inflammatory traits, that may possibly help prevent obesity. However, sex-specific effects of probiotic supplementation for targeted obesity prevention remain unknown. The aim of the current study was aimed to compare the effect of multi-probiotic preparation used in prophylactic regimen on the adiposity, profile of culturable GM and its short-chain fatty acids as well as on functional profile of phagocytes from different locations in male and female rats with monosodium glutamate (MSG)-induced obesity. Obesity was induced by neonatal MSG injections in male and female rats, who were given the multi-species probiotic during juvenile and adult developmental stages. Culturable fecal and mucosa-associated microbiota of the intestine were examined using selective diagnostic media. Short-chain fatty acid profile in fecal samples was determined by GC-MS. Phagocyte functional profile was evaluated using flow cytometry and colorimetric methods. Probiotic supplementation after the administration of MSG prevented weight gain and fat accumulation, inflammatory phagocyte activation and alterations in GM in female rats. In male MSG-injected rats, probiotic supplementation restricted but did not prevent weight gain and fat deposition, alleviated but did not prevent systemic inflammation, prevented the alterations in GM, but with residual imbalance in the ratio of obligate anaerobic to facultative anaerobic bacteria. Our findings emphasize the necessity of sex-centered approaches to the prophylactic use of probiotics in obesity in the context of predictive preventive and personalized medicine.
Collapse
Affiliation(s)
- Mariia Rudyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine,Corresponding author.
| | - Yevheniia Hurmach
- Bogomolets National Medical University, 13, T. Shevchenko Blvd, Kyiv, 01601, Ukraine
| | - Tetiana Serhiichuk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Iryna Akulenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Tetiana Berehova
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Liudmyla Ostapchenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| |
Collapse
|
44
|
Adipose Tissue and Adipose-Tissue-Derived Cell Therapies for the Treatment of the Face and Hands of Patients Suffering from Systemic Sclerosis. Biomedicines 2023; 11:biomedicines11020348. [PMID: 36830886 PMCID: PMC9953720 DOI: 10.3390/biomedicines11020348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/28/2023] Open
Abstract
Adipose tissue is recognized as a valuable source of cells with angiogenic, immunomodulatory, reparative and antifibrotic properties and emerged as a therapeutic alternative for the regeneration and repair of damaged tissues. The use of adipose-tissue-based therapy is expanding in autoimmune diseases, particularly in Systemic Sclerosis (SSc), a disease in which hands and face are severely affected, leading to disability and a decrease in quality of life. Combining the advantage of an abundant supply of fat tissue and a high abundance of stem/stromal cells, fat grafting and adipose tissue-derived cell-based therapies are attractive therapeutic options in SSc. This review aims to synthesize the evidence to determine the effects of the use of these biological products for face and hands treatment in the context of SSc. This highlights several points: the need to use relevant effectiveness criteria taking into account the clinical heterogeneity of SSc in order to facilitate assessment and comparison of innovative therapies; second, it reveals some impacts of the disease on fat-grafting success; third, an important heterogeneity was noticed regarding the manufacturing of the adipose-derived products and lastly, it shows a lack of robust evidence from controlled trials comparing adipose-derived products with standard care.
Collapse
|
45
|
Cruciani S, Delitala AP, Cossu ML, Ventura C, Maioli M. Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment. Int J Mol Sci 2023; 24:2310. [PMID: 36768633 PMCID: PMC9916844 DOI: 10.3390/ijms24032310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Obesity is a complex worldwide disease, characterized by an abnormal or excessive fat accumulation. The onset of this pathology is generally linked to a complex network of interactions among genetic and environmental factors, aging, lifestyle, and diets. During adipogenesis, several regulatory mechanisms and transcription factors are involved. As fat cells grow, adipose tissue becomes increasingly large and dysfunctional, losing its endocrine function, secreting pro-inflammatory cytokines, and recruiting infiltrating macrophages. This long-term low-grade systemic inflammation results in insulin resistance in peripheral tissues. In this review we describe the main mechanisms involved in adipogenesis, from a physiological condition to obesity. Current therapeutic strategies for the management of obesity and the related metabolic syndrome are also reported.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
| | | | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, 07100 Sassari, Italy
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering, Eldor Lab, Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via di Corticella 183, 40128 Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
46
|
Ren B, Chang Y, Liu R, Xiao F, Xu J, Li L, Li T, Ruan Z, Bao Y, Lin J, Zhou J, Liao W, Pan Z, Xu H, Tian J, Cai L, Zheng XX. Clinical phase I/II trial of SVF therapy for cartilage regeneration: A cellular therapy with novel 3D MRI imaging for evaluating chondral defect of knee osteoarthritis. Front Cell Dev Biol 2023; 11:1106279. [PMID: 36743417 PMCID: PMC9892457 DOI: 10.3389/fcell.2023.1106279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Background: The clinical applications of stromal vascular fraction (SVF) therapy for osteoarthritis (OA) have attracted academic and clinical attention. However, data of the effects of stromal vascular fraction therapy on regeneration of degenerated cartilage are limited in the literature. Meanwhile, there is a great need for a simple and non-invasive evaluation method to analyze the changes of joint cartilage qualitatively and quantitatively in clinical trials. This study entitled "stromal vascular fraction Therapy for Human Knee Osteoarthritis" was registered in ClinicalTrial.gov # NCT05019378. Materials and Methods: We designed and conducted a single center, open labeled clinical phase I/II study, and 6 osteoarthritis patients with both knee cartilage defect I-II were enrolled in this study. The two knees of each patient were randomly assigned to autologous stromal vascular fraction treatment group or non-treatment control group to evaluate the safety and therapeutic effect of stromal vascular fraction therapy for human knee osteoarthritis. We have also established a novel protocol to provide 3D MRI imaging for human knee cartilage enabling us to qualitatively and quantitatively evaluate cartilage degeneration and regeneration in this study. Results: The qualitative and quantitative evaluation of 3D Magnetic Resonance Imaging (MRI) imaging of knee cartilage demonstrated that the stromal vascular fraction therapy reduced the cartilage defects; and significant increase of cartilage value both in defect cartilage area and whole cartilage area of treated group and significant increase of thickness and area of both femoral and tibia cartilage in vertical sections of the stromal vascular fraction treated Group at 12 and 24 W post treatment in cartilage defect I-II osteoarthritis patients. Conclusion: This clinical phase I/II study indicated that stromal vascular fraction therapy is a safe clinical procedure and provided evidence that the stromal vascular fraction therapy significantly facilitated cartilage regeneration, opening the opportunity to a phase III trial investigating authentic efficacy of the procedure. This study is the first qualitative and quantitative evaluation of the efficacy of autologous stromal vascular fraction cellular therapy on cartilage regeneration. Through early and definite diagnosis of knee osteoarthritis patients, and providing safe and efficient therapy to facilitate cartilage regeneration, we will be able to control or reverse cartilage degeneration and completely change the epidemiology of osteoarthritis worldwide.
Collapse
Affiliation(s)
- Bin Ren
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiqiang Chang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ruolan Liu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Xiao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Xu
- Plastic Surgery, General Hospital of the People’s Liberation Army, Beijing, China
| | - Lingsong Li
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Tao Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhao Ruan
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yigui Bao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junbing Lin
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junying Zhou
- Clinic Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenyu Pan
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lin Cai
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China,*Correspondence: Lin Cai, ; Xin Xiao Zheng,
| | - Xin Xiao Zheng
- Department of Plastic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States,Transplantation Medical Center, Zhongnan Hospital of Wuhan University, Wuhan, China,*Correspondence: Lin Cai, ; Xin Xiao Zheng,
| |
Collapse
|
47
|
Hybrid Stromal Vascular Fraction (Hybrid-SVF): A New Paradigm in Mechanical Regenerative Cell Processing. Plast Reconstr Surg Glob Open 2022; 10:e4702. [PMID: 36601591 PMCID: PMC9803457 DOI: 10.1097/gox.0000000000004702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023]
Abstract
Enzymatic digestion of extracellular matrix (ECM) from lipoaspirate is the conventional form of harvesting stromal vascular fraction (SVF) called enzymatically digested SVF (E-SVF). Mechanical SVF (M-SVF) isolation has emerged as an alternative method, but it has also some limitations in terms of lower cell viability and diminished cell counts. To enhance the SVF qualitatively and quantitatively, we propose a novel concept called "hybrid-SVF," in which we combine M-SVF with the concentrated parts of adipose tissue after centrifugation, which is called stromal vascular matrix (SVM). Methods Hybrid-SVF injection was applied as an adjunctive therapy to fat grafting in 88 patients and 11 samples were evaluated in the laboratory for cell count, viability and cell activity. Results Experimental results determined that SVM part showed higher cellular activity. SVM and M-SVF showed higher cellular potency than E-SVF. Clinically, none of the patients required an additional session for fat grafting since there was no significant graft resorption. However, seven patients asked for further volume augmentation due to their individual preferences. No major complication was encountered. Conclusions The usage of hybrid-SVF has a very high regenerative potential due to the ECM support and exceptionally high cell yield in addition to preserved cell potency. Although there are ongoing studies focusing on optimizing cell counts and further clinical applications, we believe that our preliminary results might create a paradigm shift in the area of regenerative fat grafting.
Collapse
|
48
|
Al‐kharboosh R, Perera JJ, Bechtle A, Bu G, Quinones‐Hinojosa A. Emerging point-of-care autologous cellular therapy using adipose-derived stromal vascular fraction for neurodegenerative diseases. Clin Transl Med 2022; 12:e1093. [PMID: 36495120 PMCID: PMC9736801 DOI: 10.1002/ctm2.1093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative disorders are characterized by the gradual decline and irreversible loss of cognitive functions and CNS structures. As therapeutic recourse stagnates, neurodegenerative diseases will cost over a trillion dollars by 2050. A dearth of preventive and regenerative measures to hinder regression and enhance recovery has forced patients to settle for traditional therapeutics designed to manage symptoms, leaving little hope for a cure. In the last decade, pre-clinical animal models and clinical investigations in humans have demonstrated the safety and promise of an emerging cellular product from subcutaneous fat. The adipose-derived stromal vascular fraction (SVF) is an early intervention and late-stage novel 'at point' of care cellular treatment, demonstrating improvements in clinical applications for Multiple Sclerosis, Alzheimer's disease, and Parkinson's disease. SVF is a heterogeneous fraction of cells forming a robust cellular ecosystem and serving as a novel and valuable source of point-of-care autologous cell therapy, providing an easy-to-access population that we hypothesize can mediate repair through 'bi-directional' communication in response to pathological cues. We provide the first comprehensive review of all pre-clinical and clinical findings available to date and highlight major challenges and future directions. There is a greater medical and economic urgency to innovate and develop novel cellular therapy solutions that enable the repair and regeneration of neuronal tissue that has undergone irreversible and permanent damage.
Collapse
Affiliation(s)
- Rawan Al‐kharboosh
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida,Department of Regenerative SciencesMayo Clinic Graduate SchoolRochesterMinnesota,Department of NeurosurgeryMayo ClinicJacksonvilleFlorida
| | | | | | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida
| | | |
Collapse
|
49
|
Aratikatla A, Maffulli N, Rodriguez HC, Gupta M, Potty AG, Gupta A. Allogenic Perinatal Tissue for Musculoskeletal Regenerative Medicine Applications: A Systematic Review. Biomedicines 2022; 10:3173. [PMID: 36551929 PMCID: PMC9775213 DOI: 10.3390/biomedicines10123173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/26/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Producing tremendous amounts of stress and financial burden on the global patient population and healthcare systems around the world, most current modalities of treatment for musculoskeletal ailments often do not address the etiopathogenetic causes of these disorders. Regenerative medicine for musculoskeletal disorders relies on orthobiologics derived from either allogenic or autologous sources. Multiple drawbacks are associated with autologous sources, including donor-site morbidity, a dearth of studies, and variability in both patient reported and clinical/functional outcomes. On the other hand, allogenic sources address several of these concerns, and continue to be a suitable source of mesenchymal stem cells (MSCs). This review qualitatively reports both the preclinical and clinical outcomes of publications studying the applications of umbilical cord (-derived Wharton's jelly), amniotic suspension allograft, amniotic membrane, and amniotic fluid in musculoskeletal medicine. A systematic review was conducted utilizing the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines on studies published between January 2010 and October 2022 that used allogeneic perinatal tissues. Further randomized controlled clinical studies are necessary to properly evaluate the safety and efficacy of these tissues in orthopedic surgery.
Collapse
Affiliation(s)
- Adarsh Aratikatla
- The Royal College of Surgeons in Ireland, D02 YN77 Dublin 2, Ireland
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, 84084 Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Ortopedica” Department, Hospital of Salerno, 84124 Salerno, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent ST5 5BG, UK
| | - Hugo C. Rodriguez
- Department of Orthopaedic Surgery, Larkin Community Hospital, South Miami, FL 33143, USA
| | - Manu Gupta
- Polar Aesthetics Dental & Cosmetic Centre, Noida 201301, UP, India
| | - Anish G. Potty
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
| | - Ashim Gupta
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
- Future Biologics, Lawrenceville, GA 30043, USA
- BioIntegrate, Lawrenceville, GA 30043, USA
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, UP, India
- Regenerative Orthopaedics, Noida 201301, UP, India
| |
Collapse
|
50
|
Cittadini E, Brucculeri AM, Quartararo F, Vaglica R, Miceli V, Conaldi PG. Stem cell therapy in the treatment of organic and dysfunctional endometrial pathology. Minerva Obstet Gynecol 2022; 74:504-515. [PMID: 34851073 DOI: 10.23736/s2724-606x.21.04919-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Intrauterine adhesions caused by postpartum curettage, spontaneous abortions, interrupted pregnancies, endometrial ablations, infections and inflammations, can lead to a loss of endometrial function, with consequent hypomenorrhea and infertility in women of reproductive age. In a non-negligible percentage of cases, the available surgical methods and hormone therapy, with sequential administration of estrogen and progesterone, are ineffective. In fact, severe damage to the basal layer of the endometrium causes the loss of endometrial cell precursors and leads to the failure of regeneration of the functional layer to which the endometrium is cyclically exposed. Today, many researchers are evaluating the use of stem cells of different origins as a potential therapy to restore endometrial function. METHODS Our interest has been focused on adipose-derived stromal/stem cells (ADSCs) obtained by collecting subcutaneous adipose tissue and subsequently treating it with the MilliGraft® method. This procedure produces a cell suspension, the stromal vascular fraction (SVF), which includes ADSCs and soluble factors such as proteins and extracellular vesicles (exosomes). The SVF thus obtained was characterized in its cellular composition and its functional factors. Our clinical protocol for the future use of adipose tissue in endometrial regeneration in its different phases is presented. RESULTS The data obtained, even though they still require further support and implementation, show the regenerative properties of SVF obtained from adipose tissue using a mechanical method. CONCLUSIONS These findings can contribute to the development of cell therapies using stem cells of different derivations which are increasingly being utilized in the treatment of endometrial lesions from adherent or dysfunctional pathologies.
Collapse
Affiliation(s)
- Ettore Cittadini
- Fondazione per gli Studi sulla Riproduzione Umana, Clinica Candela, Palermo, Italy -
| | - Anna M Brucculeri
- Fondazione per gli Studi sulla Riproduzione Umana, Clinica Candela, Palermo, Italy
| | - Fabrizio Quartararo
- Fondazione per gli Studi sulla Riproduzione Umana, Clinica Candela, Palermo, Italy
| | - Roberto Vaglica
- Fondazione per gli Studi sulla Riproduzione Umana, Clinica Candela, Palermo, Italy
| | | | | |
Collapse
|