1
|
McSweeney JE, Yong LY, Goddard NV, Wong JK. Does Secondary Mechanical Manipulation of Lipoaspirate Enhance the Vasculogenic Potential of Fat Grafts? A Systematic Review. Ann Plast Surg 2024; 93:389-396. [PMID: 39150855 DOI: 10.1097/sap.0000000000004048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024]
Abstract
BACKGROUND Fat grafting is a highly versatile option in the reconstructive armamentarium but with unpredictable retention rates and outcomes. The primary outcome of this systematic review was to assess whether secondary mechanically processed lipoaspirate favorably enhances the vasculogenic potential of fat grafts when compared to unprocessed lipoaspirate or fat grafts prepared using centrifugation alone. The secondary outcome was to assess the evidence around graft retention and improved outcomes when comparing the aforementioned groups. METHODS A search on MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials was conducted up to February 2022. All human and animal research, which provided a cross-comparison between unprocessed, centrifuged, secondary mechanically fragmented (SMF) or secondary mechanically disrupted (SMD) fat grafts, was included. RESULTS Thirty-one full texts were included. Vasculogenic potential was assessed by quantification of angiogenic growth factors and cellular composition. Cellular composition of mesenchymal stem cells, perivascular stem cells, and endothelial progenitor cells was quantified by fluorescence activated cell sorting (FACS) analysis. Fat graft volume retention rates and fat grafting to aid wound healing were assessed. Although the presence of industry-funded studies and inadequate reporting of methodological data in some studies were sources of bias, data showed SMF grafts contain an enriched pericyte population with increased vascular endothelial growth factor (VEGF) secretion. Animal studies indicate that SMD grafts may increase rates of fat graft retention and wound closure compared to centrifuged grafts; however, clinical studies are yet to show similar results. CONCLUSIONS In this systematic review, we were able to conclude that the existing literature suggests mechanically processing fat, whether it be through fragmentation or disruption, improves vasculogenic potential by enhancing angiogenic growth factor and relevant vascular progenitor cell levels. Whilst in vivo animal studies are scarce, the review findings suggest that secondary mechanically processed fat enhances fat graft retention and can aid with wound healing. Further clinical studies are required to assess potential differences in human studies.
Collapse
Affiliation(s)
- Jared Ethan McSweeney
- From the Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, M13 9PL, UK
| | | | | | | |
Collapse
|
2
|
Suryawan IGR, Pikir BS, Rantam FA, Ratri AK, Nugraha RA. Hypoxic Preconditioning Promotes Survival of Human Adipose Derived Mesenchymal Stem Cell. F1000Res 2024; 10:843. [PMID: 38938689 PMCID: PMC11208860 DOI: 10.12688/f1000research.55351.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 06/29/2024] Open
Abstract
Background: Contributing factors for improved survival of human adipocytes mesenchymal stem cells (h-AMSCs) cultured through hypoxia preconditioning, in example apoptosis inhibition involving BCL2 and HSP27 expression, trigger signal expression (VEGF), SCF expression, OCT-4 expression, and CD44+ expression. The objective if this study was to explain the mechanism and role of hypoxic preconditioning and the optimal duration of hypoxic preconditioning exposure to improve survival of h-AMSCs. Methods: An experimental laboratory explorative study ( in vitro) with hypoxic preconditioning in h-AMSCs cultures. This research was conducted through four stages. First, isolation of h-AMSCs culture from adipose tissue of patients. Second, the characterization of h-AMSCs from adipose tissue by phenotype (flowcytometry) through CD44+, CD90+ and CD45-expression before being pre-conditioned for hypoxic treatment. Third, the hypoxic preconditioning in h-AMSCs culture ( in vitro) was performed with an oxygen concentration of 1% for 24, 48 and 72 hours. Fourth, observation of survival from h-AMSCs culture was tested on the role of CD44+, VEGF, SCF, OCT-4, BCL2, HSP27 with Flowcytometry and apoptotic inhibition by Tunnel Assay method. Results: The result of regression test showed that time difference had an effect on VEGF expression ( p<0.001; β=-0.482) and hypoxia condition also influenced VEGF expression ( p<0.001; β=0.774). The result of path analysis showed that SCF had effect on OCT-4 expression ( p<0.001; β=0.985). The regression test results showed that time effects on HSP27 expression ( p<0.001; β=0.398) and hypoxia precondition also affects HSP27 expression ( p<0.001; β=0.847). Pathway analysis showed that BCL2 expression inhibited apoptosis ( p=0.030; β=-0.442) and HSP27 expression also inhibited apoptosis ( p<0,001; β=-0.487). Conclusion: Hypoxic preconditioning of h-AMSC culture has proven to increase the expression of VEGF, SCF, OCT-4, and BCL2 and HSP27. This study demonstrated and explained the existence of a new mechanism of increased h-AMSC survival in cultures with hypoxic preconditioning (O2 1%) via VEGF, SCF, OCT-4, BCL2, and HSP 27.
Collapse
Affiliation(s)
- I Gde Rurus Suryawan
- Cardiology and Vascular Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - Budi Susetyo Pikir
- Cardiology and Vascular Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - Fedik Abdul Rantam
- Virology and Immunology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - Anudya Kartika Ratri
- Cardiology and Vascular Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - Ricardo Adrian Nugraha
- Cardiology and Vascular Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| |
Collapse
|
3
|
Wystrychowski G, Simka-Lampa K, Witkowska A, Sobecko E, Skubis-Sikora A, Sikora B, Wojtyna E, Golda A, Gwizdek K, Wróbel M, Sędek Ł, Górczyńska-Kosiorz S, Szweda-Gandor N, Trautsolt W, Francuz T, Kruszniewska-Rajs C, Gola J. Selected microRNA Expression and Protein Regulator Secretion by Adipose Tissue-Derived Mesenchymal Stem Cells and Metabolic Syndrome. Int J Mol Sci 2024; 25:6644. [PMID: 38928349 PMCID: PMC11204268 DOI: 10.3390/ijms25126644] [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: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The role of adipose mesenchymal stem cells (Ad-MSCs) in metabolic syndrome remains unclear. We aimed to assess the expression of selected microRNAs in Ad-MSCs of non-diabetic adults in relation to Ad-MSC secretion of protein regulators and basic metabolic parameters. Ten obese, eight overweight, and five normal weight subjects were enrolled: 19 females and 4 males; aged 43.0 ± 8.9 years. Ad-MSCs were harvested from abdominal subcutaneous fat. Ad-MSC cellular expressions of four microRNAs (2-ΔCt values) and concentrations of IL-6, IL-10, VEGF, and IGF-1 in the Ad-MSC-conditioned medium were assessed. The expressions of miR-21, miR-122, or miR-192 did not correlate with clinical parameters (age, sex, BMI, visceral fat, HOMA-IR, fasting glycemia, HbA1c, serum lipids, CRP, and eGFR). Conversely, the expression of miR-155 was lowest in obese subjects (3.69 ± 2.67 × 10-3 vs. 7.07 ± 4.42 × 10-3 in overweight and 10.25 ± 7.05 × 10-3 in normal weight ones, p = 0.04). The expression of miR-155 correlated inversely with BMI (sex-adjusted r = -0.64; p < 0.01), visceral adiposity (r = -0.49; p = 0.03), and serum CRP (r = -0.63; p < 0.01), whereas it correlated positively with serum HDL cholesterol (r = 0.51; p = 0.02). Moreover, miR-155 synthesis was associated marginally negatively with Ad-MSC secretion of IGF-1 (r = -0.42; p = 0.05), and positively with that of IL-10 (r = 0.40; p = 0.06). Ad-MSC expression of miR-155 appears blunted in visceral obesity, which correlates with Ad-MSC IGF-1 hypersecretion and IL-10 hyposecretion, systemic microinflammation, and HDL dyslipidemia. Ad-MSC studies in metabolic syndrome should focus on miR-155.
Collapse
Affiliation(s)
| | - Klaudia Simka-Lampa
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | | | - Ewelina Sobecko
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Aleksandra Skubis-Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Bartosz Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Ewa Wojtyna
- Institute of Medical Sciences, University of Opole, 45-040 Opole, Poland;
| | - Agnieszka Golda
- Alfamed General Practice, 41-100 Siemianowice Slaskie, Poland;
| | - Katarzyna Gwizdek
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marta Wróbel
- Department of Internal Medicine, Diabetology and Cardiometabolic Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Sylwia Górczyńska-Kosiorz
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Nikola Szweda-Gandor
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Wanda Trautsolt
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| | - Joanna Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| |
Collapse
|
4
|
Lauvrud AT, Giraudo MV, Wiberg R, Wiberg M, Kingham PJ, Brohlin M. The influence of xeno-free culture conditions on the angiogenic and adipogenic differentiation properties of adipose tissue-derived stem cells. Regen Ther 2024; 26:901-910. [PMID: 39822342 PMCID: PMC11736170 DOI: 10.1016/j.reth.2024.09.013] [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] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 01/19/2025] Open
Abstract
Introduction Before performing cell therapy clinical trials, it is important to understand how cells are influenced by different growth conditions and to find optimal xeno-free medium formulations. In this study we have investigated the properties of adipose tissue-derived stem cells (ASCs) cultured under xeno-free conditions. Methods Human lipoaspirate samples were digested to yield the stromal vascular fraction cells which were then seeded in i) Minimum Essential Medium-α (MEM-α) supplemented with 10 % (v/v) fetal bovine serum (FBS), ii) MEM-α supplemented with 2 % (v/v) human platelet lysate (PLT) or iii) PRIME-XV MSC expansion XSFM xeno-free, serum free medium (XV). Flow cytometry for ASCs markers CD73, CD90 and CD105 together with the putative pericyte marker CD146 was performed. Growth rates were monitored over multiple passages and adipogenic differentiation performed at early and expanded passage culture. Growth factor gene expression was analyzed and an in vitro angiogenesis assay performed. Results Cells in FBS and PLT grew at similar rates whereas the cells cultured in XV medium proliferated significantly faster up to 60 days in culture. All cultures were >98 % positive for CD73, CD90 and CD105, whereas CD146 expression was significantly higher in XV cells. Adipogenic differentiation was most pronounced in cells which had been cultured in XV medium whilst cells grown in PLT were inferior compared with cells from the FBS cultures. IGF1 gene expression was highest in cells cultured in PLT whilst cells grown in XV medium showed 10-fold lower expression compared with FBS cells. In contrast, HGF gene expression was 90-fold greater in cells cultured in XV medium compared with those cultured in FBS. Conditioned medium from XV cultured cells showed the most angiogenic activity, inducing the greatest endothelial cell network formation and maturation. Conclusion Culture under different conditions alters the ASCs characteristics. Since cells cultured in XV medium showed the best adipogenic and angiogenic profile this might be a preferred medium formulation for preparing cells required for reconstructive surgical applications such as cell-assisted fat grafting.
Collapse
Affiliation(s)
- Anne Therese Lauvrud
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Maria Vittoria Giraudo
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Rebecca Wiberg
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Mikael Wiberg
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Paul J. Kingham
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Maria Brohlin
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
5
|
Ren H, Zhang L, Zhang X, Yi C, Wu L. Specific lipid magnetic sphere sorted CD146-positive bone marrow mesenchymal stem cells can better promote articular cartilage damage repair. BMC Musculoskelet Disord 2024; 25:253. [PMID: 38561728 PMCID: PMC10983655 DOI: 10.1186/s12891-024-07381-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND The characteristics and therapeutic potential of subtypes of bone marrow mesenchymal stem cells (BMSCs) are largely unknown. Also, the application of subpopulations of BMSCs in cartilage regeneration remains poorly characterized. The aim of this study was to explore the regenerative capacity of CD146-positive subpopulations of BMSCs for repairing cartilage defects. METHODS CD146-positive BMSCs (CD146 + BMSCs) were sorted by self-developed CD146-specific lipid magnetic spheres (CD146-LMS). Cell surface markers, viability, and proliferation were evaluated in vitro. CD146 + BMSCs were subjected to in vitro chondrogenic induction and evaluated for chondrogenic properties by detecting mRNA and protein expression. The role of the CD146 subpopulation of BMSCs in cartilage damage repair was assessed by injecting CD146 + BMSCs complexed with sodium alginate gel in the joints of a mouse cartilage defect model. RESULTS The prepared CD146-LMS had an average particle size of 193.7 ± 5.24 nm, an average potential of 41.9 ± 6.21 mv, and a saturation magnetization intensity of 27.2 Am2/kg, which showed good stability and low cytotoxicity. The sorted CD146 + BMSCs highly expressed stem cell and pericyte markers with good cellular activity and cellular value-added capacity. Cartilage markers Sox9, Collagen II, and Aggrecan were expressed at both protein and mRNA levels in CD146 + BMSCs cells after chondrogenic induction in vitro. In a mouse cartilage injury model, CD146 + BMSCs showed better function in promoting the repair of articular cartilage injury. CONCLUSION The prepared CD146-LMS was able to sort out CD146 + BMSCs efficiently, and the sorted subpopulation of CD146 + BMSCs had good chondrogenic differentiation potential, which could efficiently promote the repair of articular cartilage injury, suggesting that the sorted CD146 + BMSCs subpopulation is a promising seed cell for cartilage tissue engineering.
Collapse
Affiliation(s)
- Hanru Ren
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Lele Zhang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Xu Zhang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Chengqing Yi
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China.
| | - Lianghao Wu
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China.
| |
Collapse
|
6
|
Picoli CDC, Birbrair A, Li Z. Pericytes as the Orchestrators of Vasculature and Adipogenesis. Genes (Basel) 2024; 15:126. [PMID: 38275607 PMCID: PMC10815550 DOI: 10.3390/genes15010126] [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: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Pericytes (PCs) are located surrounding the walls of small blood vessels, particularly capillaries and microvessels. In addition to their functions in maintaining vascular integrity, participating in angiogenesis, and regulating blood flow, PCs also serve as a reservoir for multi-potent stem/progenitor cells in white, brown, beige, and bone marrow adipose tissues. Due to the complex nature of this cell population, the identification and characterization of PCs has been challenging. A comprehensive understanding of the heterogeneity of PCs may enhance their potential as therapeutic targets for metabolic syndromes or bone-related diseases. This mini-review summarizes multiple PC markers commonly employed in lineage-tracing studies, with an emphasis on their contribution to adipogenesis and functions in different adipose depots under diverse metabolic conditions.
Collapse
Affiliation(s)
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Madison, WI 53706, USA;
| | - Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA;
| |
Collapse
|
7
|
Strauß S, Diemer M, Bucan V, Kuhbier JW, Asendorf T, Vogt PM, Schlottmann F. Spider silk enhanced tissue engineering of cartilage tissue: Approach of a novel bioreactor model using adipose derived stromal cells. J Appl Biomater Funct Mater 2024; 22:22808000241226656. [PMID: 38253568 DOI: 10.1177/22808000241226656] [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] [Indexed: 01/24/2024] Open
Abstract
Human cartilage tissue remains a challenge for the development of therapeutic options due to its poor vascularization and reduced regenerative capacities. There are a variety of research approaches dealing with cartilage tissue engineering. In addition to different biomaterials, numerous cell populations have been investigated in bioreactor-supported experimental setups to improve cartilage tissue engineering. The concept of the present study was to investigate spider silk cocoons as scaffold seeded with adipose-derived stromal cells (ASC) in a custom-made bioreactor model using cyclic axial compression to engineer cartilage-like tissue. For chemical induction of differentiation, BMP-7 and TGF-β2 were added and changes in cell morphology and de-novo tissue formation were investigated using histological staining to verify chondrogenic differentiation. By seeding spider silk cocoons with ASC, a high colonization density and cell proliferation could be achieved. Mechanical induction of differentiation using a newly established bioreactor model led to a more roundish cell phenotype and new extracellular matrix formation, indicating a chondrogenic differentiation. The addition of BMP-7 and TGF-β2 enhanced the expression of cartilage specific markers in immunohistochemical staining. Overall, the present study can be seen as pilot study and valuable complementation to the published literature.
Collapse
Affiliation(s)
- Sarah Strauß
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Maximilian Diemer
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Jörn W Kuhbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
- Department of Plastic, Aesthetic and Hand Surgery, Helios Klinikum Hildesheim, Hildesheim, Germany
| | - Tomke Asendorf
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Frederik Schlottmann
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Smolinska A, Bzinkowska A, Rybkowska P, Chodkowska M, Sarnowska A. Promising Markers in the Context of Mesenchymal Stem/Stromal Cells Subpopulations with Unique Properties. Stem Cells Int 2023; 2023:1842958. [PMID: 37771549 PMCID: PMC10533301 DOI: 10.1155/2023/1842958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023] Open
Abstract
The heterogeneity of the mesenchymal stem/stromal cells (MSCs) population poses a challenge to researchers and clinicians, especially those observed at the population level. What is more, the lack of precise evidences regarding MSCs developmental origin even further complicate this issue. As the available evidences indicate several possible pathways of MSCs formation, this diverse origin may be reflected in the unique subsets of cells found within the MSCs population. Such populations differ in specialization degree, proliferation, and immunomodulatory properties or exhibit other additional properties such as increased angiogenesis capacity. In this review article, we attempted to identify such outstanding populations according to the specific surface antigens or intracellular markers. Described groups were characterized depending on their specialization and potential therapeutic application. The reports presented here cover a wide variety of properties found in the recent literature, which is quite scarce for many candidates mentioned in this article. Even though the collected information would allow for better targeting of specific subpopulations in regenerative medicine to increase the effectiveness of MSC-based therapies.
Collapse
Affiliation(s)
- Agnieszka Smolinska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Aleksandra Bzinkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Magdalena Chodkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| |
Collapse
|
9
|
Ishida Y, Mabuchi Y, Naraoka Y, Hisamatsu D, Akazawa C. Conservation of Markers and Stemness in Adipose Stem and Progenitor Cells between Cattle and Other Species. Int J Mol Sci 2023; 24:11908. [PMID: 37569284 PMCID: PMC10418360 DOI: 10.3390/ijms241511908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Adipose stem and progenitor cells (ASPCs) have been isolated from humans and animals for use in regenerative medicine and therapy. However, knowledge of ASPCs in other species is limited. Particularly, ASPCs in livestock are expected to enhance the fat content and meat composition. In this study, we isolated bovine ASPCs using cell surface markers. Specifically, we focused on ASPC markers in humans and experimental animals, namely CD26, CD146, and CD54. Stromal vascular fraction cells from bovine fat were separated using flow cytometry before primary culture. We evaluated the self-renewal and adipogenic potential of each fraction. We identified four cell populations: CD26-CD146+CD54+, CD26-CD146+CD54-, CD26-CD146-, and CD26+CD146-. Among them, the CD26-CD146+ fraction, particularly CD54+, demonstrated the properties of preadipocytes (PreAs), characterized by slow proliferation and a high adipogenic capacity. In conclusion, we could collect and characterize possible PreAs as CD26-CD146+CD54+ or CD26-CD146+CD54-, which are expected for in vitro bovine adipogenic assays in the future.
Collapse
Affiliation(s)
- Yuki Ishida
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.I.); (Y.M.); (Y.N.); (D.H.)
| | - Yo Mabuchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.I.); (Y.M.); (Y.N.); (D.H.)
- Department of Clinical Regenerative Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Yuna Naraoka
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.I.); (Y.M.); (Y.N.); (D.H.)
| | - Daisuke Hisamatsu
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.I.); (Y.M.); (Y.N.); (D.H.)
| | - Chihiro Akazawa
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.I.); (Y.M.); (Y.N.); (D.H.)
| |
Collapse
|
10
|
Skrypnik D, Skrypnik K, Suliburska J, Bogdański P. Leptin-VEGF crosstalk in excess body mass and related disorders: A systematic review. Obes Rev 2023:e13575. [PMID: 37230803 DOI: 10.1111/obr.13575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/17/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
By 2030, it is expected that a billion people will have suffer from obesity. Adipose tissue synthesizes leptin, an adipokine that affects cardiovascular risk. Leptin intensifies the synthesis of vascular endothelial growth factor (VEGF). Our study reviews recent reports on leptin-VEGF crosstalk in obesity and related disorders. PubMed, Web of Science, Scopus, and Google Scholar were searched. One hundred and one articles involving human, animal, and in vitro research were included. In vitro studies show the crucial role of interaction between endothelial cells and adipocytes and hypoxia as a factor that intensifies leptin's effects on VEGF. Leptin-VEGF crosstalk promotes the progression of cancer. The animal research reveal that a high-fat diet enhances leptin and VEGF crosstalk. Genetic and epigenetic mechanisms and procreator-offspring programming may be involved in leptin-VEGF crosstalk. Some female-specific characteristics of leptin-VEGF relation in obesity were observed. The human studies have shown that increased leptin and VEGF synthesis and leptin-VEGF crosstalk are factors linking obesity with elevated cardiovascular risk. The studies of the last 10 years documented a range of significant aspects of leptin-VEGF crosstalk specific for obesity and related disorders, shedding new light on the link between obesity and increased cardiovascular risk.
Collapse
Affiliation(s)
- Damian Skrypnik
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Skrypnik
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Suliburska
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
11
|
Liu M, Shang Y, Liu N, Zhen Y, Chen Y, An Y. Strategies to Improve AFT Volume Retention After Fat Grafting. Aesthetic Plast Surg 2023; 47:808-824. [PMID: 36316460 DOI: 10.1007/s00266-022-03088-y] [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: 07/15/2022] [Accepted: 08/28/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Autologous fat grafting has gained increasing popularity used in plastic surgery as a strategy to improve functional and aesthetic outcome. However, variable augmentation results have concerned surgeons in that volume loss of grafted fat reported fluctuates unsteadily. AIM An optimal technique that clinically maximizes the long-term survival rate of transplantation is in urgent need to be identified. METHOD The PubMed/MEDLINE database was queried to search for animal and human studies published through March of 2022 with search terms related to adipose grafting encompassing liposuction, adipose graft viability, processing technique, adipose-derived stem cell, SVF and others. RESULTS 45 in vivo studies met inclusion criteria. The principal of ideal processing technique is effective purification of fat and protection of tissue viability, such as gauze rolling and washing-filtration devices. Cell-assisted lipotransfer including SVF, SVF-gel and ADSCs significantly promotes graft retention via differentiation potential and paracrine manner. ADSCs induce polarization of macrophages to regulate inflammatory response, mediate extracellular matrix remodeling and promote endothelial cell migration and sprouting, and differentiate into adipocytes to replace necrotic cells, providing powerful evidence for the benefits and efficacy of cell-assisted lipotransfer. CONCLUSION Based on the current evidence, the best strategy can not be decided. Cell-assisted lipotransfer has great potential for use in regenerative medicine. But so far mechanically prepared SVF-gel is conducive to clinical promotion. PRP as endogenous growth factor sustained-release material shows great feasibility. LEVEL OF EVIDENCE IV This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Meiling Liu
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yujia Shang
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Na Liu
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Youbai Chen
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
12
|
Schlottmann F, Bucan V, Strauß S, Koop F, Vogt PM, Mett TR. Influence of Tamoxifen on Different Biological Pathways in Tumorigenesis and Transformation in Adipose-Derived Stem Cells, Mammary Cells and Mammary Carcinoma Cell Lines—An In Vitro Study. Cells 2022; 11:cells11172733. [PMID: 36078139 PMCID: PMC9454616 DOI: 10.3390/cells11172733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/17/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022] Open
Abstract
Breast carcinoma is one of the most common malignant tumors in women. In cases of hormone-sensitive cells, tamoxifen as an anti-estrogenic substance is a first line medication in the adjuvant setting. The spectrum of autologous breast reconstructions ranges from fat infiltrations to complex microsurgical procedures. The influence of adipose-derived stem cells (ASC) on the tumor bed and a possibly increased recurrence rate as a result are critically discussed. In addition, there is currently no conclusive recommendation regarding tamoxifen-treated patients and autologous fat infiltrations. The aim of the present study was to investigate the effect of tamoxifen on the gene expression of a variety of genes involved in tumorigenesis, cell growth and transformation. Mammary epithelial cell line and mammary carcinoma cell lines were treated with tamoxifen in vitro as well as co-cultured with ASC. Gene expression was quantified by PCR arrays and showed increased expression in the mammary carcinoma cell lines with increasing time of treatment and concentration of tamoxifen. The data presented can be considered as an addition to the controversial discussion on the relationship between ASC and breast carcinoma cells. Further studies are needed to quantify the in vivo interaction of ASC and mammary carcinoma cells and to conclusively assess the impact of tamoxifen in reconstructive cases with fat grafting.
Collapse
|
13
|
Wu M, Huang Y, Zhu Q, Zhu X, Xue L, Xiong J, Chen Y, Wu C, Guo Y, Li Y, Wu M, Wang S. Adipose tissue and ovarian aging: Potential mechanism and protective strategies. Ageing Res Rev 2022; 80:101683. [PMID: 35817297 DOI: 10.1016/j.arr.2022.101683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/29/2022] [Accepted: 07/05/2022] [Indexed: 11/01/2022]
Abstract
Ovarian aging occurs approximately 10 years prior to the natural age-associated functional decline of other organ systems. With the increase of life expectancy worldwide, ovarian aging has gradually become a key health problem among women. Therefore, understanding the causes and molecular mechanisms of ovarian aging is very essential for the inhibition of age-related diseases and the promotion of health and longevity in women. Recently, studies have revealed an association between adipose tissue (AT) and ovarian aging. Alterations in the function and quantity of AT have profound consequences on ovarian function because AT is central for follicular development, lipid metabolism, and hormonal regulation. Moreover, the interplay between AT and the ovary is bidirectional, with ovary-derived signals directly affecting AT biology. In this review, we summarize the current knowledge of the complex molecular mechanisms controlling the crosstalk between the AT and ovarian aging, and further discuss how therapeutic targeting of the AT can delay ovarian aging.
Collapse
Affiliation(s)
- Meng Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yibao Huang
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Qingqing Zhu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Xiaoran Zhu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Liru Xue
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Chen
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Chuqing Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yican Guo
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yinuo Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Mingfu Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| |
Collapse
|
14
|
Zhang L, Sun Y, Zhang XX, Liu YB, Sun HY, Wu CT, Xiao FJ, Wang LS. Comparison of CD146 +/- mesenchymal stem cells in improving premature ovarian failure. Stem Cell Res Ther 2022; 13:267. [PMID: 35729643 PMCID: PMC9209844 DOI: 10.1186/s13287-022-02916-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are a heterogeneous group of subpopulations with differentially expressed surface markers. CD146 + MSCs correlate with high therapeutic and secretory potency. However, their therapeutic efficacy and mechanisms in premature ovarian failure (POF) have not been explored. METHODS The umbilical cord (UC)-derived CD146 +/- MSCs were sorted using magnetic beads. The proliferation of MSCs was assayed by dye670 staining and flow cytometry. A mouse POF model was established by injection of cyclophosphamide and busulfan, followed by treatment with CD146 +/- MSCs. The therapeutic effect of CD146 +/- MSCs was evaluated based on body weight, hormone levels, follicle count and reproductive ability. Differential gene expression was identified by mRNA sequencing and validated by RT-PCR. The lymphocyte percentage was detected by flow cytometry. RESULTS CD146 +/- MSCs had similar morphology and surface marker expression. However, CD146 + MSCs exhibited a significantly stronger proliferation ability. Gene profiles revealed that CD146 + MSCs had a lower levels of immunoregulatory factor expression. CD146 + MSCs exhibited a stronger ability to inhibit T cell proliferation. CD146 +/- MSCs treatment markedly restored FSH and E2 hormone secretion level, reduced follicular atresia, and increased sinus follicle numbers in a mouse POF model. The recovery function of CD146 + MSCs in a reproductive assay was slightly improved than that of CD146 - MSCs. Ovary mRNA sequencing data indicated that UC-MSCs therapy improved ovarian endocrine locally, which was through PPAR and cholesterol metabolism pathways. The percentages of CD3, CD4, and CD8 lymphocytes were significantly reduced in the POF group compared to the control group. CD146 + MSCs treatment significantly reversed the changes in lymphocyte percentages. Meanwhile, CD146 - MSCs could not improve the decrease in CD4/8 ratio induced by chemotherapy. CONCLUSION UC-MSCs therapy improved premature ovarian failure significantly. CD146 +/- MSCs both had similar therapeutic effects in repairing reproductive ability. CD146 + MSCs had advantages in modulating immunology and cell proliferation characteristics.
Collapse
Affiliation(s)
- Lin Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Laboratory of Molecular Diagnosis and Regenerative Medicine, Medical Research Center, The Affiliate Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Yang Sun
- Laboratory of Molecular Diagnosis and Regenerative Medicine, Medical Research Center, The Affiliate Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Xiao-Xu Zhang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, Medical Research Center, The Affiliate Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Yu-Bin Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Hui-Yan Sun
- Yanda Medical Research Institute, Hebei Yanda Hospital, Sanhe, 065201, Hebei Province, People's Republic of China
| | - Chu-Tse Wu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Feng-Jun Xiao
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Li-Sheng Wang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, Medical Research Center, The Affiliate Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
| |
Collapse
|
15
|
Comparative study of mouse adipose- and bone marrow mesenchymal stem cells in diabetic model with critical limb ischemia. Cell Tissue Bank 2022; 23:923-936. [PMID: 35590084 DOI: 10.1007/s10561-022-10007-7] [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: 12/25/2021] [Accepted: 04/13/2022] [Indexed: 11/02/2022]
Abstract
The aim of this research is to compare the capabilities of Adipose tissue mesenchymal stem cells (AT-MSCs) and bone marrow mesenchymal stem cells (BM-MSCs) in the treatment of diabetic male mice with CLI model. Supernatants were collected from C57BL/6 mice isolated AT-MSCs and BM-MSCs, afterward their effects on human umbilical vein endothelial (HUVEC) migration potential were evaluated. Diabetes mellitus type 1 was induced by streptozotocin injection. Diabetic mice with CLI model were divided into three groups and injected with AT-MSCs, BM-MSCs, or PBS then the efficacy of them was assessed. Survival of MSCs was analysed by SRY-specific gene. The conditioned medium of AT-MSCs and BM-MSCs stimulated HUVECs migration and the donor cells were detected till 21 day in two groups. BM-MSCs and AT-MSCs improved significantly functional recovery and ischemia damage. Neovascularization in ischemic muscle was significantly higher in mice treated with AT-MSCs and BM-MSCs and they improved muscle regeneration. In vivo and in vitro findings show that AT-MSCs and BM-MSCs transplantation could be proposed as a promising therapy to promote angiogenesis and muscle regeneration through secretion of proangiogenic factors, cytokines and growth factors in diabetic mice with CLI model wherein blood supply is insufficient and disrupted.
Collapse
|
16
|
Peng Q, Ren G, Xuan Z, Duda M, Pennisi CP, Porsborg SR, Fink T, Zachar V. Distinct Dominant Lineage from In Vitro Expanded Adipose-Derived Stem Cells (ASCs) Exhibits Enhanced Wound Healing Properties. Cells 2022; 11:cells11071236. [PMID: 35406800 PMCID: PMC8998068 DOI: 10.3390/cells11071236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/16/2022] Open
Abstract
It has been suggested that immunophenotypically defined lineages within the in vitro expanded adipose-derived stem cell (ASC) may play a beneficial role from the perspective of a personalized intervention. Therefore, to better understand the implications of different surface marker profiles for the functionality, we set out to examine the evolution of ASC-variants based on the co-expression of five bright or eight dim epitopes. At passages P1, P4, and P8, the co-localization of five bright markers (CD73, CD90, CD105, CD166, and CD201), or eight dim markers (CD34, CD36, CD200, CD248, CD271, CD274, CD146, and the Stro-1), was investigated by flow cytometry. Selected subpopulations were isolated using the fluorescence-activated cells sorting from the cryopreserved P4 and analyzed in terms of proliferative and clonogenic properties, trilineage differentiation, and wound healing potential. Only two of the dim epitopes were found in representative subpopulations (SP), and from the P4 onwards, two major combinations featuring the CD274+ (SP1) or the CD274+ CD146+ (SP2) emerged. Upon sorting and growth, both subpopulations assumed new but highly similar clonal profiles, consisting of the CD274+ CD146+ and the CD274+ CD146+ CD248+ phenotypes. The functional analysis revealed that the SP2 surpassed SP1 and the unfractionated cells regarding the growth rate, clonogenic activity, and the wound closure and endothelial tube formation potential. The surface epitopes may be considered a tool to enrich specific functionality and thus improve therapeutic outcomes in dedicated circumstances.
Collapse
|
17
|
Deleon NMD, Adem S, Lavin CV, Abbas DB, Griffin M, King ME, Borrelli MR, Patel RA, Fahy EJ, Lee D, Shen AH, Momeni A, Longaker MT, Wan DC. Angiogenic CD34+CD146+ adipose-derived stromal cells augment recovery of soft tissue after radiotherapy. J Tissue Eng Regen Med 2021; 15:1105-1117. [PMID: 34582109 DOI: 10.1002/term.3253] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/26/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022]
Abstract
Radiation therapy is effective for cancer treatment but may also result in collateral soft tissue contracture, contour deformities, and non-healing wounds. Autologous fat transfer has been described to improve tissue architecture and function of radiation-induced fibrosis and these effects may be augmented by enrichment with specific adipose-derived stromal cells (ASCs) with enhanced angiogenic potential. CD34+CD146+, CD34+CD146-, or CD34+ unfractionated human ASCs were isolated by flow cytometry and used to supplement human lipoaspirate placed beneath the scalp of irradiated mice. Volume retention was followed radiographically and fat grafts as well as overlying soft tissue were harvested after eight weeks for histologic and biomechanical analyses. Radiographic evaluation revealed the highest volume retention in fat grafts supplemented with CD34+CD146+ ASCs, and these grafts were also found to have greater histologic integrity than other groups. Irradiated skin overlying CD34+CD146+ ASC-enriched grafts was significantly more vascularized than other treatment groups, had significantly less dermal thickness and collagen deposition, and the greatest improvement in fibrillin staining and return of elasticity. Radiation therapy obliterates vascularity and contributes to scarring and loss of tissue function. ASC-enrichment of fat grafts with CD34+CD146+ ASCs not only enhances fat graft vascularization and retention, but also significantly promotes improvement in overlying radiation-injured soft tissue. This regenerative effect on skin is highly promising for patients with impaired wound healing and deformities following radiotherapy.
Collapse
Affiliation(s)
- Nestor M Diaz Deleon
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Sandeep Adem
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Christopher V Lavin
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Darren B Abbas
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle Griffin
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Megan E King
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Mimi R Borrelli
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Ronak A Patel
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Evan J Fahy
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel Lee
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Abra H Shen
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Arash Momeni
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C Wan
- Department of Surgery, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
18
|
Scioli MG, Storti G, Bielli A, Sanchez M, Scimeca M, Gimble JM, Cervelli V, Orlandi A. CD146 expression regulates osteochondrogenic differentiation of human adipose-derived stem cells. J Cell Physiol 2021; 237:589-602. [PMID: 34287857 DOI: 10.1002/jcp.30506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 01/12/2023]
Abstract
Tissue engineering aims to develop innovative approaches to repair tissue defects. The use of adipose-derived stem cells (ASCs) in tissue regeneration was extensively investigated for osteochondrogenesis. Among the ASC population, ASCs expressing the CD146 were demonstrated to be multipotent and considered as perivascular stem cells, although the functional role of CD146 expression in these cells remains unclear. Herein, we investigated the influence of CD146 expression on osteochondrogenic differentiation of ASCs. Our results showed that, in two-dimensional culture systems, sorted CD146+ ASCs proliferated less and displayed higher adipogenic and chondrogenic potential than CD146- ASCs. The latter demonstrated a higher osteogenic capacity. Besides this, CD146+ ASCs in three-dimensional Matrigel/endothelial growth medium (EGM) cultures showed the highest angiogenic capability. When cultured in three-dimensional collagen scaffolds, CD146+ ASCs showed a spontaneous chondrogenic differentiation, further enhanced by the EGM medium's addition. Finally, CD146- ASCs seeded on hexafluoroisopropanol silk scaffolds displayed a greater spontaneous osteogenetic capacity. Altogether, these findings demonstrated a functional and relevant influence of CD146 expression in ASC properties and osteochondrogenic commitment. Exploiting the combination of specific differentiation properties of ASC subpopulations and appropriate culture systems could represent a promising strategy to improve the efficacy of new regenerative therapies.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Alessandra Bielli
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Sanchez
- Major Equipments and Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Manuel Scimeca
- Anatomic Pathology, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Jeffrey M Gimble
- Department of Pharmacology, Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.,Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, Tirana, Albania
| |
Collapse
|
19
|
Lauvrud AT, Gümüscü R, Wiberg R, Brohlin M, Kelk P, Wiberg M, Kingham PJ. Water jet-assisted lipoaspiration and Sepax cell separation system for the isolation of adipose stem cells with high adipogenic potential. J Plast Reconstr Aesthet Surg 2021; 74:2759-2767. [PMID: 33994109 DOI: 10.1016/j.bjps.2021.03.025] [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: 09/15/2020] [Revised: 02/05/2021] [Accepted: 03/11/2021] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Water jet-assisted liposuction has gained popularity due to favourable fat grafting outcomes. In this study, we compared stem cells obtained from fat isolated with manual or the water jet-assisted procedure. METHODS Liposuction of abdominal fat was performed using the two methods on each donor (n = 10). Aspirate samples were collagenase digested and the isolated cells seeded in vitro prior to proliferation, adipogenic differentiation and angiogenic activity analyses. RESULTS Cells from either procedure proliferated at similar rates and exhibited a similar colony-forming ability. The cells expressed stem cell markers CD73, CD90 and CD105. In the water jet cell preparations, there were higher numbers of cells expressing CD146. Robust adipogenic differentiation was observed in cultures expanded from both manual and water jet lipoaspirates. Gene analysis showed higher expression of the adipocyte markers aP2 and GLUT4 in the adipocyte-differentiated water jet cell preparations, and ELISA indicated increased secretion of adiponectin from these cells. Both cell groups expressed vasculogenic factors and the water jet cells promoted the highest levels of in vitro angiogenesis. Given these positive results, we further characterised the water jet cells when prepared using an automated closed cell processing unit, the Sepax-2 system (Cytiva). The growth and stem cell properties of the Sepax-processed cells were similar to the standard centrifugation protocol, but there was evidence for greater adipogenic differentiation in the Sepax-processed cells. CONCLUSIONS Water jet lipoaspirates yield cells with high adipogenic potential and angiogenic activity, which may be beneficial for use in cell-assisted lipotransfers.
Collapse
Affiliation(s)
- Anne Therese Lauvrud
- Department of Integrative Medical Biology, Umeå University, Sweden; Department of Surgical and Perioperative Sciences, Umeå University, Umeå 907 37, Sweden.
| | - Rojda Gümüscü
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå 907 37, Sweden
| | - Rebecca Wiberg
- Department of Integrative Medical Biology, Umeå University, Sweden; Department of Surgical and Perioperative Sciences, Umeå University, Umeå 907 37, Sweden
| | - Maria Brohlin
- Department of Clinical Microbiology, Infection and Immunity, Umeå University, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Umeå University, Sweden
| | - Mikael Wiberg
- Department of Integrative Medical Biology, Umeå University, Sweden; Department of Surgical and Perioperative Sciences, Umeå University, Umeå 907 37, Sweden
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, Sweden
| |
Collapse
|
20
|
Zha K, Tian G, Yang Z, Sun Z, Liu S, Guo Q. [The role of CD146 in mesenchymal stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:227-233. [PMID: 33624479 DOI: 10.7507/1002-1892.202005110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To summarize the expression and role of CD146 in mesenchymal stem cells (MSCs). Methods The literature related to CD146 at home and abroad were extensively consulted, and the CD146 expression in MSCs and its function were summarized and analyzed. Results CD146 is a transmembrane protein that mediates the adhesion of cells to cells and extracellular matrix, and is expressed on the surface of various MSCs. More and more studies have shown that CD146 + MSCs have superior cell properties such as greater proliferation, differentiation, migration, and immune regulation abilities than CD146 - or unsorted MSCs, and the application of CD146 + MSCs in the treatment of specific diseases has also achieved better results. CD146 is also involved in mediating a variety of cellular signaling pathways, but whether it plays the same role in MSCs remains to be demonstrated by further experiments. Conclusion The utilization of CD146 + MSCs for tissue regeneration will be conducive to improving the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, 100853, P.R.China;Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China;School of Medicine, Nankai University, Tianjin, 300071, P.R.China
| | - Shuyun Liu
- Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China
| | - Quanyi Guo
- Institute of Orthopaedics, the First Medical Centre, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Beijing, 100853, P.R.China
| |
Collapse
|
21
|
Yi Y, Hu W, Zhao C, Wu M, Zeng H, Xiong M, Lv W, Wu Y, Zhang Q. Deciphering the Emerging Roles of Adipocytes and Adipose-Derived Stem Cells in Fat Transplantation. Cell Transplant 2021; 30:963689721997799. [PMID: 33650919 PMCID: PMC7930646 DOI: 10.1177/0963689721997799] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 01/01/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
Autologous fat transplantation is widely regarded as an increasingly popular method for augmentation or reshaping applications in soft tissue defects. Although the fat transplantation is of simple applicability, low donor site morbidity and excellent biocompatibility, the clinical unpredictability and high resorption rates of the fat grafts remain an inevitable problem. In the sites of fat transplantation, the most essential components are the adipocyte and adipose-derived stem cells (ADSCs). The survival of adipocytes is the direct factor determining fat retention. The efficacy of fat transplantation is reduced by fat absorption and fibrosis due to the inadequate blood flow, adipocyte apoptosis and fat necrosis. ADSCs, a heterogeneous mixture of cells in adipose tissue, are closely related to tissue survival. ADSCs exhibit the ability of multilineage differentiation and remarkable paracrine activity, which is crucial for graft survival. This article will review the recent existing research on the mechanisms of adipocytes and ADSCs in fat transplantation, especially including adipocyte apoptosis, mature adipocyte dedifferentiation, adipocyte browning, ADSCs adipogenic differentiation and ADSCs angiogenesis. The in-depth understanding of the survival mechanism will be extremely valuable for achieving the desired filling effects.
Collapse
Affiliation(s)
- Yi Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijie Hu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongru Zhao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zeng
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingchen Xiong
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenchang Lv
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Caponnetto F, Manini I, Bulfoni M, Zingaretti N, Miotti G, Di Loreto C, Cesselli D, Mariuzzi L, Parodi PC. Human Adipose-Derived Stem Cells in Madelung's Disease: Morphological and Functional Characterization. Cells 2020; 10:cells10010044. [PMID: 33396896 PMCID: PMC7824042 DOI: 10.3390/cells10010044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Madelung Disease (MD) is a syndrome characterized by the accumulation of aberrant symmetric adipose tissue deposits. The etiology of this disease is yet to be elucidated, even though the presence of comorbidities, either genetic or environmental, has been reported. For this reason, establishing an in vitro model for MD is considered crucial to get insights into its physiopathology. We previously established a protocol for isolation and culture of stem cells from diseased tissues. Therefore, we isolated human adipose-derived stem cells (ASC) from MD patients and compared these cells with those isolated from healthy subjects in terms of surface phenotype, growth kinetic, adipogenic differentiation potential, and molecular alterations. Moreover, we evaluated the ability of the MD-ASC secretome to affect healthy ASC. The results reported a difference in the growth kinetic and surface markers of MD-ASC compared to healthy ASC but not in adipogenic differentiation. The most commonly described mitochondrial mutations were not observed. Still, MD-ASC secretome was able to shift the healthy ASC phenotype to an MD phenotype. This work provides evidence of the possibility of exploiting a patient-based in vitro model for better understanding MD pathophysiology, possibly favoring the development of novel target therapies.
Collapse
Affiliation(s)
- Federica Caponnetto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Correspondence: ; Tel.: +39-04-3255-9412
| | - Ivana Manini
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Michela Bulfoni
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
| | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| | - Giovanni Miotti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| | - Carla Di Loreto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Daniela Cesselli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Laura Mariuzzi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Pier Camillo Parodi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| |
Collapse
|
23
|
Borrelli MR, Patel RA, Blackshear C, Vistnes S, Diaz Deleon NM, Adem S, Shen AH, Sokol J, Momeni A, Nguyen D, Longaker MT, Wan DC. CD34+CD146+ adipose-derived stromal cells enhance engraftment of transplanted fat. Stem Cells Transl Med 2020; 9:1389-1400. [PMID: 32543083 PMCID: PMC7581443 DOI: 10.1002/sctm.19-0195] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 04/24/2020] [Accepted: 05/24/2020] [Indexed: 12/16/2022] Open
Abstract
Fat grafting is a surgical technique able to reconstruct and regenerate soft tissue. The adipose‐derived stromal cells (ASCs) within the stromal vascular fraction are believed to drive these beneficial effects. ASCs are increasingly recognized to be a heterogeneous group, comprised of multiple stem and progenitor subpopulations with distinct functions. We hypothesized the existence of an ASC subpopulation with enhanced angiogenic potential. Human ASCs that were CD34+CD146+, CD34+CD146−, or CD34+ unfractionated (UF) were isolated by flow cytometry for comparison of expression of proangiogenic factors and endothelial tube‐forming potential. Next, lipoaspirate was enriched with either CD34+CD146+, CD34+CD146−, CD34+ UF ASCs, or was not enriched, and grafted beneath the scalp skin of immunodeficient CD‐1 Nude mice (10 000 cells/200 μL/graft). Fat retention was monitored radiographically more than 8 weeks and fat grafts were harvested for histological assessment of quality and vascularization. The CD34+CD146+ subpopulation comprised ~30% of ASCs, and exhibited increased expression of vascular endothelial growth factor and angiopoietin‐1 compared to CD34+CD146− and CD34+ UF ASCs, and increased expression of fibroblast growth factor‐2 compared to CD34+CD146− ASCs. The CD34+CD146+ subpopulation exhibited enhanced induction of tube‐formation compared to CD34+CD146− ASCs. Upon transplantation, fat enriched CD34+CD146+ ASCs underwent less resorption and had improved histologic quality and vascularization. We have identified a subpopulation of CD34+ ASCs with enhanced angiogenic effects in vitro and in vivo, likely mediated by increased expression of potent proangiogenic factors. These findings suggest that enriching lipoaspirate with CD34+CD146+ ASCs may enhance fat graft vascularization and retention in the clinical setting.
Collapse
Affiliation(s)
- Mimi R Borrelli
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ronak A Patel
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Charles Blackshear
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stephanie Vistnes
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Nestor M Diaz Deleon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Sandeep Adem
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Abra H Shen
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jan Sokol
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dung Nguyen
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
24
|
Son Y, Cox JM, Stevenson JL, Cooper JA, Paton CM. Angiopoietin-1 protects 3T3-L1 preadipocytes from saturated fatty acid–induced cell death. Nutr Res 2020; 76:20-28. [DOI: 10.1016/j.nutres.2020.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/25/2019] [Accepted: 02/07/2020] [Indexed: 12/14/2022]
|
25
|
Vu NB, Phi LT, Dao TTT, Le HTN, Ta VT, Pham PV. Adipose derived stem cell transplantation is better than bone marrow mesenchymal stem cell transplantation in treating hindlimb ischemia in mice. BIOMEDICAL RESEARCH AND THERAPY 2019. [DOI: 10.7603/s40730-016-0046-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
26
|
Chu DT, Nguyen Thi Phuong T, Tien NLB, Tran DK, Minh LB, Thanh VV, Gia Anh P, Pham VH, Thi Nga V. Adipose Tissue Stem Cells for Therapy: An Update on the Progress of Isolation, Culture, Storage, and Clinical Application. J Clin Med 2019; 8:E917. [PMID: 31247996 PMCID: PMC6678927 DOI: 10.3390/jcm8070917] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/10/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue stem cells (ASCs), known as multipotent stem cells, are most commonly used in the clinical applications in recent years. Adipose tissues (AT) have the advantage in the harvesting, isolation, and expansion of ASCs, especially an abundant amount of stem cells compared to bone marrow. ASCs can be found in stromal vascular fractions (SVF) which are easily obtained from the dissociation of adipose tissue. Both SVFs and culture-expanded ASCs exhibit the stem cell characteristics such as differentiation into multiple cell types, regeneration, and immune regulators. Therefore, SVFs and ASCs have been researched to evaluate the safety and benefits for human use. In fact, the number of clinical trials on ASCs is going to increase by years; however, most trials are in phase I and II, and lack phase III and IV. This systemic review highlights and updates the process of the harvesting, characteristics, isolation, culture, storage, and application of ASCs, as well as provides further directions on the therapeutic use of ASCs.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam.
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam
| | - Le Bui Minh
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh St., Ward 13, District 4, Ho Chi Minh City 700000, Vietnam
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Pham Gia Anh
- Oncology Department, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Vu Thi Nga
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam.
| |
Collapse
|
27
|
Damous LL, de Carvalho AETS, Nakamuta JS, Shiroma ME, Louzada ACS, Soares-Jr JM, Krieger JE, Baracat EC. Cell-free therapy with the secretome of adipose tissue-derived stem cells in rats' frozen-thawed ovarian grafts. Stem Cell Res Ther 2018; 9:323. [PMID: 30463630 PMCID: PMC6249760 DOI: 10.1186/s13287-018-1054-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/29/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023] Open
Abstract
The use of secretome may be a new strand of cell therapy, which is equal to or even superior to the injection of live cells, called cell-free therapy. In ovarian transplantation, this approach may be a therapeutic possibility for the ovarian graft in hypoxia. We designed the present study to evaluate whether the cell-free therapy with the secretome of adipose tissue-derived stem cells (ASCs) in rat frozen-thawed ovarian grafts could protect a graft against ischemic injury. A single dose of rat ASCs secretome or vehicle was injected into the bilateral frozen-thawed ovaries of 18 adult female rats immediately after an autologous transplant. Nine animals were used to control the cryopreservation protocol and were evaluated before and after the cryopreservation process. Daily vaginal smears were performed for estrous cycle evaluation until euthanasia on postoperative day 30. Follicle viability by trypan blue, graft morphology by HE, and apoptosis by TUNEL and cleaved-caspase-3 were assessed. No differences were found with respect to estrous cycle resumption and follicle viability (p > 0.05). However, compared with the vehicle-treated grafts, the morphology of the secretome-treated grafts was impaired, showing reduced follicular population and increased apoptosis (p < 0.05). ASC secretome impaired the rat frozen-thawed ovarian graft from ischemic injury. However, more studies are needed to evaluate the factors involved and the possibility of applying the secretome in scaffolds to optimize its use.
Collapse
Affiliation(s)
- Luciana Lamarão Damous
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil. .,Baturite St, 120. Ap 91., Aclimação, São Paulo, 01530-030, Brazil.
| | - Ana Elisa Teófilo Saturi de Carvalho
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Juliana Sanajotti Nakamuta
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Marcos Eiji Shiroma
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - Andressa Cristina Sposato Louzada
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - José Maria Soares-Jr
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Edmund C Baracat
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| |
Collapse
|
28
|
Zhang R, Li R, Feng Q, Zhi L, Li Z, Xu YO, Lin Y. Expression profiles and associations of FGF1 and FGF10 with intramuscular fat in Tibetan chicken. Br Poult Sci 2018; 59:613-617. [PMID: 30259763 DOI: 10.1080/00071668.2018.1507018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- R. Zhang
- College of Life Sciences, Hubei Normal University, Huangshi, China
| | - R. Li
- Reproductive and Endocrine Laboratory, Chengdu Woman-Child Central Hospital, Chengdu, China
| | - Q. Feng
- Department of Otorhinolaryngology Head and Neck Surgery, Hospital of Guangxi Medical University, Nanning, China
| | - L. Zhi
- College of Life Sciences, Hubei Normal University, Huangshi, China
| | - Z. Li
- College of Life Science and Technology, Southwest University for Nationalities, Chengdu, China
| | - Y.-O. Xu
- College of Life Science and Technology, Southwest University for Nationalities, Chengdu, China
| | - Y. Lin
- College of Life Science and Technology, Southwest University for Nationalities, Chengdu, China
| |
Collapse
|
29
|
Combined therapy for critical limb ischaemia: Biomimetic PLGA microcarriers potentiates the pro-angiogenic effect of adipose tissue stromal vascular fraction cells. J Tissue Eng Regen Med 2018; 12:1363-1373. [DOI: 10.1002/term.2667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 03/28/2018] [Indexed: 11/07/2022]
|
30
|
Gabrielli M, Romero DG, Martini CN, Raiger Iustman LJ, Vila MDC. MCAM knockdown impairs PPARγ expression and 3T3-L1 fibroblasts differentiation to adipocytes. Mol Cell Biochem 2018; 448:299-309. [PMID: 29468504 DOI: 10.1007/s11010-018-3334-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/15/2018] [Indexed: 11/25/2022]
Abstract
We investigated for the first time the expression of melanoma cell adhesion molecule (MCAM) and its involvement in the differentiation of 3T3-L1 fibroblasts to adipocytes. We found that MCAM mRNA increased subsequent to the activation of the master regulator of adipogenesis, PPARγ, and this increase was maintained in the mature adipocytes. On the other hand, MCAM knockdown impaired differentiation and induction of PPARγ as well as expression of genes activated by PPARγ. However, events that precede and are necessary for early PPARγ activation, such as C/EBPβ induction, β-catenin downregulation, and ERK activation, were not affected in the MCAM knockdown cells. In keeping with this, the increase in PPARγ mRNA that precedes MCAM induction was not altered in the knockdown cells. In conclusion, our findings suggest that MCAM is a gene upregulated and involved in maintaining PPARγ induction in the late but not in the early stages of 3T3-L1 fibroblasts adipogenesis.
Collapse
Affiliation(s)
- Matías Gabrielli
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
- Universidad de Buenos Aires, CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Damián G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Claudia N Martini
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
| | - Laura Judith Raiger Iustman
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
- Universidad de Buenos Aires, CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - María Del C Vila
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina.
- Universidad de Buenos Aires, CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
31
|
Pericytes in Veterinary Species: Prospective Isolation, Characterization and Tissue Regeneration Potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1109:67-77. [DOI: 10.1007/978-3-030-02601-1_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Esteves CL, Donadeu FX. Pericytes and their potential in regenerative medicine across species. Cytometry A 2017; 93:50-59. [PMID: 28941046 DOI: 10.1002/cyto.a.23243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/15/2017] [Accepted: 08/25/2017] [Indexed: 12/17/2022]
Abstract
The discovery that pericytes are in vivo counterparts of Mesenchymal Stem/Stromal Cells (MSCs) has placed these perivascular cells in the research spotlight, bringing up hope for a well-characterized cell source for clinical applications, alternative to poorly defined, heterogeneous MSCs preparations currently in use. Native pericytes express typical MSC markers and, after isolation by fluorescence-activated cell sorting, display an MSC phenotype in culture. These features have been demonstrated in different species, including humans and horses, the main targets of regenerative treatments. Significant clinical potential of pericytes has been shown by transplantation of human cells into rodent models of tissue injury, and it is hoped that future studies will demonstrate clinical potential in veterinary species. Here, we provide an overview of the current knowledge on pericytes across different species including humans, companion and large animal models, in relation to their identification in different body tissues, methodology for prospective isolation, characterization, and potential for tissue regeneration. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- C L Esteves
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| | - F X Donadeu
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| |
Collapse
|
33
|
Gettler BC, Zakhari JS, Gandhi PS, Williams SK. Formation of Adipose Stromal Vascular Fraction Cell-Laden Spheroids Using a Three-Dimensional Bioprinter and Superhydrophobic Surfaces. Tissue Eng Part C Methods 2017; 23:516-524. [PMID: 28665236 DOI: 10.1089/ten.tec.2017.0056] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The therapeutic infusion of adipose-derived stromal vascular fraction (SVF) cells for the treatment of multiple diseases, has progressed to numerous human clinical trials; however, the often poor retention of the cells following implantation remains a common drawback of direct cell injection. One solution to cellular retention at the injection site has been the use of biogels to encapsulate cells within a microenvironment before and upon implantation. The current study utilized three-dimensional bioprinting technology to evaluate the ability to form SVF cell-laden spheroids with collagen I as a gel-forming biomatrix. A superhydrophobic surface was created to maintain the bioprinted structures in a spheroid shape. A hydrophilic disc was printed onto the hydrophobic surface to immobilize the spheroids during the gelation process. Conditions for the automated rapid formation of SVF cell-laden spheroids were explored, including time/pressure relationships for spheroid extrusion during bioprinting. The formed spheroids maintain SVF viability in both static culture and dynamic spinner culture. Spheroids also undergo a time-dependent contraction with the retention of angiogenic sprout phenotype over the 14-day culture period. The use of a biphilic surface exhibiting both superhydrophobicity to maintain the spheroid shape and a hydrophilicity to immobilize the spheroid during gel formation produces SVF cell-laden spheroids that can be immediately transplanted for therapeutic applications.
Collapse
Affiliation(s)
- Brian C Gettler
- Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| | - Joseph S Zakhari
- Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| | - Piyani S Gandhi
- Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| | - Stuart K Williams
- Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| |
Collapse
|
34
|
Brett E, Zielins ER, Chin M, Januszyk M, Blackshear CP, Findlay M, Momeni A, Gurtner GC, Longaker MT, Wan DC. Isolation of CD248-expressing stromal vascular fraction for targeted improvement of wound healing. Wound Repair Regen 2017; 25:414-422. [PMID: 28464475 DOI: 10.1111/wrr.12542] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
Wound healing remains a global issue of disability, cost, and health. Addition of cells from the stromal vascular fraction (SVF) of adipose tissue has been shown to increase the rate of full thickness wound closure. This study aimed to investigate the angiogenic mechanisms of CD248+ SVF cells in the context of full thickness excisional wounds. Single cell transcriptional analysis was used to identify and cluster angiogenic gene-expressing cells, which was then correlated with surface marker expression. SVF cells isolated from human lipoaspirate were FACS sorted based on the presence of CD248. Cells were analyzed for angiogenic gene expression and ability to promote microvascular tubule formation in vitro. Following this, 6mm full thickness dermal wounds were created on the dorsa of immunocompromised mice and then treated with CD248+, CD248-, or unsorted SVF cells delivered in a pullalan-collagen hydrogel or the hydrogel alone. Wounds were measured every other day photometrically until closure. Wounds were also evaluated histologically at 7 and 14 days post-wounding and when fully healed to assess for reepithelialization and development of neovasculature. Wounds treated with CD248+ cells healed significantly faster than other treatment groups, and at 7 days, had quantitatively more reepithelialization. Concurrently, immunohistochemistry of CD31 revealed a much higher presence of vascularity in the CD248+ SVF cells treated group at the time of healing and at 14 days post-op, consistent with a pro-angiogenic effect of CD248+ cells in vivo. Therefore, using CD248+ pro-angiogenic cells obtained from SVF presents a viable strategy in wound healing by promoting increased vessel growth in the wound.
Collapse
Affiliation(s)
- Elizabeth Brett
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Elizabeth R Zielins
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Monica Chin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Charles P Blackshear
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael Findlay
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
35
|
Xie G, Swiderska-Syn M, Jewell ML, Machado MV, Michelotti GA, Premont RT, Diehl AM. Loss of pericyte smoothened activity in mice with genetic deficiency of leptin. BMC Cell Biol 2017; 18:20. [PMID: 28427343 PMCID: PMC5399438 DOI: 10.1186/s12860-017-0135-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/06/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Obesity is associated with multiple diseases, but it is unclear how obesity promotes progressive tissue damage. Recovery from injury requires repair, an energy-expensive process that is coupled to energy availability at the cellular level. The satiety factor, leptin, is a key component of the sensor that matches cellular energy utilization to available energy supplies. Leptin deficiency signals energy depletion, whereas activating the Hedgehog pathway drives energy-consuming activities. Tissue repair is impaired in mice that are obese due to genetic leptin deficiency. Tissue repair is also blocked and obesity enhanced by inhibiting Hedgehog activity. We evaluated the hypothesis that loss of leptin silences Hedgehog signaling in pericytes, multipotent leptin-target cells that regulate a variety of responses that are often defective in obesity, including tissue repair and adipocyte differentiation. RESULTS We found that pericytes from liver and white adipose tissue require leptin to maintain expression of the Hedgehog co-receptor, Smoothened, which controls the activities of Hedgehog-regulated Gli transcription factors that orchestrate gene expression programs that dictate pericyte fate. Smoothened suppression prevents liver pericytes from being reprogrammed into myofibroblasts, but stimulates adipose-derived pericytes to become white adipocytes. Progressive Hedgehog pathway decay promotes senescence in leptin-deficient liver pericytes, which, in turn, generate paracrine signals that cause neighboring hepatocytes to become fatty and less proliferative, enhancing vulnerability to liver damage. CONCLUSIONS Leptin-responsive pericytes evaluate energy availability to inform tissue construction by modulating Hedgehog pathway activity and thus, are at the root of progressive obesity-related tissue pathology. Leptin deficiency inhibits Hedgehog signaling in pericytes to trigger a pericytopathy that promotes both adiposity and obesity-related tissue damage.
Collapse
Affiliation(s)
- Guanhua Xie
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
| | - Marzena Swiderska-Syn
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
- Current address: Medical University of South Carolina, Charleston, SC 29425 USA
| | - Mark L. Jewell
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
| | - Mariana Verdelho Machado
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
- Current address: Santa Maria Hospital, University of Lisbon, Lisbon, Portugal
| | - Gregory A. Michelotti
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
- Current address: Metabolon Inc, Research Triangle Park, NC 27709 USA
| | - Richard T. Premont
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
| | - Anna Mae Diehl
- Department of Medicine, Division of Gastroenterology, Duke University, 905 S. LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710 USA
| |
Collapse
|
36
|
Pericytes: The Role of Multipotent Stem Cells in Vascular Maintenance and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:69-86. [PMID: 29282647 DOI: 10.1007/5584_2017_138] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Blood vessels consist of an inner endothelial cell layer lining the vessel wall and perivascular pericytes, also known as mural cells, which envelop the vascular tube surface. Pericytes have recently been recognized for their central role in blood vessel formation. Pericytes are multipotent cells that are heterogeneous in their origin, function, morphology and surface markers. Similar to other types of stem cells, pericytes act as a repair system in response to injury by maintaining the structural integrity of blood vessels. Several studies have shown that blood vessels lacking pericytes become hyperdilated and haemorrhagic, leading to vascular complications ranging from diabetic retinopathy to embryonic death. The role of pericytes is not restricted to the formation and development of the vasculature: they have been shown to possess stem cell-like characteristics and may differentiate into cell types from different lineages. Recent discoveries regarding the contribution of pericytes to tumour metastasis and the maintenance of tumour vascular supply and angiogenesis have led researchers to propose targeting pericytes with anti-angiogenic therapies. In this review, we will examine the different physiological roles of pericytes, their differentiation potential, and how they interact with surrounding cells to ensure the integrity of blood vessel formation and maintenance.
Collapse
|
37
|
He J, Chen DL, Samocha-Bonet D, Gillinder KR, Barclay JL, Magor GW, Perkins AC, Greenfield JR, Yang G, Whitehead JP. Fibroblast growth factor-1 (FGF-1) promotes adipogenesis by downregulation of carboxypeptidase A4 (CPA4) - a negative regulator of adipogenesis implicated in the modulation of local and systemic insulin sensitivity. Growth Factors 2016; 34:210-216. [PMID: 28209092 DOI: 10.1080/08977194.2017.1285764] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor-1 (FGF-1) promotes differentiation of human preadipocytes into mature adipocytes via modulation of a BMP and Activin Membrane-Bound Inhibitor (BAMBI)/Peroxisome proliferator-activated receptor (PPARγ)-dependent network. Here, we combined transcriptomic and functional investigations to identify novel downstream effectors aligned with complementary analyses of gene expression in human adipose tissue to explore relationships with insulin sensitivity. RNA-Seq and qRT-PCR analysis revealed significant down-regulation of carboxypeptidase A4 (CPA4) following FGF-1 treatment or induction of differentiation of human preadipocytes in a BAMBI/PPARγ-independent manner. siRNA-mediated knockdown of CPA4 resulted in enhanced differentiation of human preadipocytes. Furthermore, expression of CPA4 in subcutaneous adipose tissue correlated negatively with indices of local and systemic (liver and muscle) insulin sensitivity. These results identify CPA4 as a negative regulator of adipogenesis that is down-regulated by FGF-1 and a putative deleterious modulator of local and systemic insulin sensitivity. Further investigations are required to define the molecular mechanism(s) involved and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Jingjing He
- a Laboratory of Animal Fat Deposition and Muscle Development , College of Animal Science and Technology, Northwest A&F University , Yangling , P.R. China
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Daniel L Chen
- c Garvan Institute of Medical Research , Sydney , Australia
| | - Dorit Samocha-Bonet
- c Garvan Institute of Medical Research , Sydney , Australia
- d Faculty of Medicine , University of New South Wales , Randwick , Australia
| | - Kevin R Gillinder
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Johanna L Barclay
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Graham W Magor
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Andrew C Perkins
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
| | - Jerry R Greenfield
- c Garvan Institute of Medical Research , Sydney , Australia
- d Faculty of Medicine , University of New South Wales , Randwick , Australia
- e Department of Endocrinology and Diabetes , St Vincent's Hospital , Sydney , Australia , and
| | - Gongshe Yang
- a Laboratory of Animal Fat Deposition and Muscle Development , College of Animal Science and Technology, Northwest A&F University , Yangling , P.R. China
| | - Jonathan P Whitehead
- b Mater Research Institute-University of Queensland, Translational Research Institute , Brisbane , Australia
- f School of Life Sciences , University of Lincoln , Lincolnshire , UK
| |
Collapse
|
38
|
Vezzani B, Pierantozzi E, Sorrentino V. Not All Pericytes Are Born Equal: Pericytes from Human Adult Tissues Present Different Differentiation Properties. Stem Cells Dev 2016; 25:1549-1558. [PMID: 27549576 DOI: 10.1089/scd.2016.0177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pericytes (PCs) have been recognized for a long time only as structural cells of the blood vessels. The identification of tight contacts with endothelial cells and the ability to interact with surrounding cells through paracrine signaling revealed additional functions of PCs in maintaining the homeostasis of the perivascular environment. PCs got the front page, in the late 1990s, after the identification and characterization of a new embryonic cell population, the mesoangioblasts, from which PCs present in the adult organism are thought to derive. From these studies, it was clear that PCs were also endowed with multipotent mesodermal abilities. Furthermore, their ability to cross the vascular wall and to reconstitute skeletal muscle tissue after systemic injection opened the way to a number of studies aimed to develop therapeutic protocols for a cell therapy of muscular dystrophy. This has resulted in a major effort to characterize pericytic cell populations from skeletal muscle and other adult tissues. Additional studies also addressed their relationship with other cells of the perivascular compartment and with mesenchymal stem cells. These data have provided initial evidence that PCs from different adult tissues might be endowed with distinctive differentiation abilities. This would suggest that the multipotent mesenchymal ability of PCs might be restrained within different tissues, likely depending on the specific cell renewal and repair requirements of each tissue. This review presents current knowledge on human PCs and highlights recent data on the differentiation properties of PCs isolated from different adult tissues.
Collapse
Affiliation(s)
- Bianca Vezzani
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| |
Collapse
|