51
|
Recent Patents Involving Stromal Vascular Fraction. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00283-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
52
|
Zhang Z, Wang J, Lin Y, Chen J, Liu J, Zhang X. Nutritional activities of luteolin in obesity and associated metabolic diseases: an eye on adipose tissues. Crit Rev Food Sci Nutr 2022; 64:4016-4030. [PMID: 36300856 DOI: 10.1080/10408398.2022.2138257] [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: 11/03/2022]
Abstract
Obesity is characterized by excessive body fat accumulation and is a high-risk factor for metabolic comorbidities, including type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular disease. In lean individuals, adipose tissue (AT) is not only an important regulatory organ for energy storage and metabolism, but also an indispensable immune and endocrine organ. The sustained energy imbalance induces adipocyte hypotrophy and hyperplasia as well as AT remodeling, accompanied by chronic low-grade inflammation and adipocytes dysfunction in AT, ultimately leading to systemic insulin resistance and ectopic lipid deposition. Luteolin is a natural flavonoid widely distributed in fruits and vegetables and possesses multifold biological activities, such as antioxidant, anticancer, and anti-inflammatory activities. Diet supplementation of this flavonoid has been reported to inhibit AT lipogenesis and inflammation as well as the ectopic lipid deposition, increase AT thermogenesis and systemic energy expenditure, and finally improve obesity and associated metabolic diseases. The purpose of this review is to reveal the nutritional activities of luteolin in obesity and its complications with emphasis on its action on AT energy metabolism, immunoregulation, and endocrine intervention.
Collapse
Affiliation(s)
- Zhixin Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jiahui Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
- Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui, China
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| |
Collapse
|
53
|
Intraoperative Administration of Adipose Stromal Vascular Fraction Does Not Improve Functional Outcomes in Young Patients with Anterior Cruciate Ligament Reconstruction. J Clin Med 2022; 11:jcm11216240. [PMID: 36362468 PMCID: PMC9654782 DOI: 10.3390/jcm11216240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Adipose stromal vascular fraction (SVF) has a versatile cellular system for biologically augmented therapies. However, there have been no clinical studies investigating the benefits of the augmentation of anterior cruciate ligament reconstruction (ACLR) with SVF. We conducted a retrospective study in assessing the effects of intraoperative SVF administration on the functional outcomes in young patients with ACLR. The enrolled patients were divided into the control group (ACLR only) and the SVF group (ACLR with SVF). The functional outcomes in both groups were assessed by the Lysholm knee scoring system, the Tegner activity scale, and the International Knee Documentation Committee (IKDC) subjective evaluation form, and compared at several time points during a 12-month follow-up. We found that the sex distribution and pre-surgery scores were similar in the two groups, whereas the mean age of the SVF group was higher than that of the control group (p = 0.046). The between-group analysis and generalized estimating equation model analysis revealed that, while patients in the SVF group significantly improved all their functional outcomes at 12 months after surgery, this improvement was not significantly different from the results of patients in the control group (Lysholm, p = 0.553; Tegner, p = 0.197; IKDC, p = 0.486). No side effects were observed in either group. We concluded that the intraoperative administration of SVF does not improve or accelerate functional recovery after ACLR in young patients.
Collapse
|
54
|
The role of patient characteristics and the effects of angiogenic therapies on the microvasculature of the meniscus: A systematic review. Knee 2022; 38:91-106. [PMID: 35964436 DOI: 10.1016/j.knee.2022.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 07/15/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Considerable interindividual variation in meniscal microvascularization has been reported. The purpose of this review was to identify which patient characteristics affect meniscal microvascularization and provide a structured overview of angiogenic therapies that influence meniscal neovascularization. METHODS A systematic literature search was undertaken using PubMed, Embase, Web of Science, Cochrane library and Emcare from inception to November 2021. Studies reporting on (1) Patient characteristics that affect meniscal microvascularization, or (2) Therapies that induce neovascularization in meniscal tissue were included. Studies were graded in quality using the Anatomical Quality Assessment (AQUA) tool. The study was registered with PROSPERO(ID:CRD42021242479). RESULTS Thirteen studies reported on patient characteristics and eleven on angiogenic therapies. The influence of Age, Degenerative knee, Gender, and Race was reported. Age is the most studied factor. The entire meniscus is vascularized around birth. With increasing age, vascularization decreases from the inner to the peripheral margin. Around 11 years, blood vessels are primarily located in the peripheral third of the menisci. There seems to be a further decrease in vascularization with increasing age in adults, yet conflicting literature exists. Degenerative changes of the knee also seem to influence meniscal vascularization, but evidence is limited. Angiogenic therapies to improve meniscal vascularization have only been studied in preclinical setting. The use of synovial flap transplantation, stem cell therapy, vascular endothelial growth factor, and angiogenin has shown promising results. CONCLUSION To decrease failure rates of meniscal repair, a better understanding of patient-specific vascular anatomy is essential. Translational clinical research is needed to investigate the clinical value of angiogenic therapies.
Collapse
|
55
|
Cao L, Xiaoming F, Zhang Q, Fang J, Chu C, Lv J, Ma Y, Lu G, Yang K, Pan R. An Optimized Method for Adipose Stromal Vascular Fraction Isolation and its Application in Fat Grafting. Aesthetic Plast Surg 2022; 46:2500-2508. [PMID: 34981156 DOI: 10.1007/s00266-021-02738-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND The stromal vascular fraction (SVF) derived from adipose tissue contains heterogeneous cell populations and has enormous potential for clinical therapy. There are two main methods for SVF isolation: enzymatic isolation and mechanical isolation, both of which have shortcomings. In this study, optimized conditions for the isolation of high-quality SVF were established, and applications in fat grafting were evaluated. METHODS Adipose tissue was chopped into small pieces and then ground into an erosive shape using a syringe. The pieces were digested with 0.15% type II collagenase for 35 min at 37 °C. After centrifugation, the pellets were resuspended in DMEM and passed through a 100-μm strainer. The filtered cells were analyzed by flow cytometry. The fat graft was enriched with isolated SVF and subcutaneously transplanted into nude mice. Three weeks after transplantation, grafts were isolated, and H&E staining, immunocytochemistry, and western blotting were conducted. RESULTS The harvested SVF cells reached > 2 × 106/ml of adipose tissue within 90 min of operation. The number of CD34+ ADSCs in our SVF pellets was > 6 × 105/ml of adipose tissue, which has the potential for differentiating into osteoblasts, adipocytes, and chondrocytes. Freshly collected adipose tissue is better for SVF isolation, and isolated SVF should also be kept at 4 °C and used as soon as possible. SVF may promote revascularization after fat grafting. The adipose tissue of an SVF co-transplanted group had an integral structure, clear capillaries, and higher VEGF expression. SVF co-transplantation inhibited adipose cell apoptosis. CONCLUSION Our study provides an efficient procedure for SVF isolation, its application in fat grafting, and possible underlying mechanisms. 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)
- Liang Cao
- Department of Acupuncture and Moxibustion, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng Xiaoming
- Department of Neurosurgery, The First Hospital of Jiaxing, Affiliated hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Qiang Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Junbiao Fang
- Department of Anesthesiology, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chunhua Chu
- Department of Acupuncture and Moxibustion, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jinlong Lv
- Department of Acupuncture and Moxibustion, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuyuan Ma
- Department of Neurosurgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Gang Lu
- Department of Neurosurgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kaichuang Yang
- Department of Neurosurgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Ruolang Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China.
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China.
| |
Collapse
|
56
|
Lee J, Park BC, Jang NY, Lee S, Cho YK, Sharma P, Byun SW, Jeon K, Jeon Y, Park U, Ro HJ, Park HR, Kim Y, Lee D, Chung S, Kim YK, Cho N. Inducing Ectopic T Cell Clusters Using Stromal Vascular Fraction Spheroid-Based Immunotherapy to Enhance Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203842. [PMID: 36058002 PMCID: PMC9534947 DOI: 10.1002/advs.202203842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/21/2022] [Indexed: 05/09/2023]
Abstract
Tertiary lymphoid structures (TLSs) provide specialized niches for immune cells, resulting in improved prognoses for patients undergoing cancer immunotherapy. Shaping TLS-like niches may improve anti-cancer immunity and overcome the current limitations of immune cell-based immunotherapy. Here, it is shown that stromal vascular fraction (SVF) from adipose tissues can enhance dendritic cell (DC)-mediated T cell immunity by inducing ectopic T lymphocyte clusters. SVF cells expanded ex vivo have phenotypes and functions similar to those of fibroblastic reticular cells in a secondary lymphoid organ, and their properties can be modulated using three-dimensional spheroid culture and coculture with DCs spiked with antigen-loaded iron oxide-zinc oxide core-shell nanoparticles. Thereby, the combination of SVF spheroids and mature DCs significantly augments T cell recruitment and retention at the injection site. This strategy elicits enhanced antigen-specific immune response and anti-tumoral immunity in mice, illustrating the potential for a novel immunotherapeutic design using SVF as a structural scaffold for TLS.
Collapse
Affiliation(s)
- Jae‐Won Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
| | - Bum Chul Park
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
- Brain Korea Center for Smart Materials and DevicesKorea UniversitySeoul02841Korea
| | - Na Yoon Jang
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Sihyeon Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Young Kyu Cho
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Prashant Sharma
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Sang Won Byun
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
| | - Kyeongseok Jeon
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Yun‐Hui Jeon
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
| | - Uni Park
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Hyo Jin Ro
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Hyo Ree Park
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Yuri Kim
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
| | - Dong‐Sup Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
| | - Seok Chung
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Young Keun Kim
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
- Brain Korea Center for Smart Materials and DevicesKorea UniversitySeoul02841Korea
| | - Nam‐Hyuk Cho
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
- Seoul National University Bundang HospitalSeongnam‐siGyeonggi‐do13620Republic of Korea
| |
Collapse
|
57
|
Freitas-Ribeiro S, Diogo GS, Oliveira C, Martins A, Silva TH, Jarnalo M, Horta R, Reis RL, Pirraco RP. Growth Factor-Free Vascularization of Marine-Origin Collagen Sponges Using Cryopreserved Stromal Vascular Fractions from Human Adipose Tissue. Mar Drugs 2022; 20:md20100623. [PMID: 36286447 PMCID: PMC9604698 DOI: 10.3390/md20100623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The successful integration of transplanted three-dimensional tissue engineering (TE) constructs depends greatly on their rapid vascularization. Therefore, it is essential to address this vascularization issue in the initial design of constructs for perfused tissues. Two of the most important variables in this regard are scaffold composition and cell sourcing. Collagens with marine origins overcome some issues associated with mammal-derived collagen while maintaining their advantages in terms of biocompatibility. Concurrently, the freshly isolated stromal vascular fraction (SVF) of adipose tissue has been proposed as an advantageous cell fraction for vascularization purposes due to its highly angiogenic properties, allowing extrinsic angiogenic growth factor-free vascularization strategies for TE applications. In this study, we aimed at understanding whether marine collagen 3D matrices could support cryopreserved human SVF in maintaining intrinsic angiogenic properties observed for fresh SVF. For this, cryopreserved human SVF was seeded on blue shark collagen sponges and cultured up to 7 days in a basal medium. The secretome profile of several angiogenesis-related factors was studied throughout culture times and correlated with the expression pattern of CD31 and CD146, which showed the formation of a prevascular network. Upon in ovo implantation, increased vessel recruitment was observed in prevascularized sponges when compared with sponges without SVF cells. Immunohistochemistry for CD31 demonstrated the improved integration of prevascularized sponges within chick chorioalantoic membrane (CAM) tissues, while in situ hybridization showed human cells lining blood vessels. These results demonstrate the potential of using cryopreserved SVF combined with marine collagen as a streamlined approach to improve the vascularization of TE constructs.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Gabriela S. Diogo
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Catarina Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Tiago H. Silva
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
- Correspondence:
| |
Collapse
|
58
|
Simão JJ, Cruz MM, Abdala FM, Bolsoni-Lopes A, Armelin-Correa L, Alonso-Vale MIC. Palmitoleic Acid Acts on Adipose-Derived Stromal Cells and Promotes Anti-Hypertrophic and Anti-Inflammatory Effects in Obese Mice. Pharmaceuticals (Basel) 2022; 15:1194. [PMID: 36297306 PMCID: PMC9609051 DOI: 10.3390/ph15101194] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 04/16/2024] Open
Abstract
Adipose tissue (AT) secretes adipokines, modulators of low-grade chronic inflammation in obesity. Molecules that induce the emergence of new and functional adipocytes in AT can alleviate or prevent inflammatory and metabolic disorders. The objective of this study was to investigate the role of palmitoleic acid (n7) in 3T3-L1 and primary pre-adipocyte differentiation and AT inflammation. C57BL/6j mice were submitted to a control or high-fat diet (HFD) for 8 weeks, and treated with n7 for 4 weeks. Mice consuming a HFD presented an increase in body weight, epididymal (Epi) fat mass, and Epi adipocytes size. N7 treatment attenuated the body weight gain and completely prevented the hypertrophy of Epi adipocytes, but not the increment in Epi mass induced by the HFD, suggesting a greater adipocytes hyperplasia in animals treated with n7. It was agreed that n7 increased 3T3-L1 proliferation and differentiation, as well as the expression of genes involved in adipogenesis, such as Cebpa, Pparg, aP2, Perilipin, and Scl2a4. Furthermore, n7 decreased the inflammatory cytokines Mcp1, Tnfa, Il6, Cxcl10, and Nos2 genes in Epi vascular stromal cells, but not in the whole AT. These findings show that n7 exerts anti-hypertrophic effects in adipocytes which influence the surrounding cells by attenuating the overexpression of pro-inflammatory cytokines triggered by a HFD.
Collapse
Affiliation(s)
- Jussara J. Simão
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Maysa M. Cruz
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Fernanda M. Abdala
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Andressa Bolsoni-Lopes
- Department of Nursing, Health Sciences Center, Federal University of Espirito Santo, Vitoria 29075-910, ES, Brazil
| | - Lucia Armelin-Correa
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Maria Isabel C. Alonso-Vale
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| |
Collapse
|
59
|
Rowe G, Heng DS, Beare JE, Hodges NA, Tracy EP, Murfee WL, LeBlanc AJ. Stromal Vascular Fraction Reverses the Age-Related Impairment in Revascularization following Injury. J Vasc Res 2022; 59:343-357. [PMID: 36075199 PMCID: PMC9780192 DOI: 10.1159/000526002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/06/2022] [Indexed: 12/31/2022] Open
Abstract
Adipose-derived stromal vascular fraction (SVF) has emerged as a potential regenerative therapy, but few studies utilize SVF in a setting of advanced age. Additionally, the specific cell population in SVF providing therapeutic benefit is unknown. We hypothesized that aging would alter the composition of cell populations present in SVF and its ability to promote angiogenesis following injury, a mechanism that is T cell-mediated. SVF isolated from young and old Fischer 344 rats was examined with flow cytometry for cell composition. Mesenteric windows from old rats were isolated following exteriorization-induced (EI) hypoxic injury and intravenous injection of one of four cell therapies: (1) SVF from young or (2) old donors, (3) SVF from old donors depleted of or (4) enriched for T cells. Advancing age increased the SVF T-cell population but reduced revascularization following injury. Both young and aged SVF incorporated throughout the host mesenteric microvessels, but only young SVF significantly increased vascular area following EI. This study highlights the effect of donor age on SVF angiogenic efficacy and demonstrates how the ex vivo mesenteric-window model can be used in conjunction with SVF therapy to investigate its contribution to angiogenesis.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA,
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA,
| | - David S Heng
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Nicholas A Hodges
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Evan P Tracy
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
60
|
Sorrell JM, Caplan AI. Heparan Sulfate: A Regulator of White Adipocyte Differentiation and of Vascular/Adipocyte Interactions. Biomedicines 2022; 10:biomedicines10092115. [PMID: 36140217 PMCID: PMC9495464 DOI: 10.3390/biomedicines10092115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 11/30/2022] Open
Abstract
White adipose tissues are major endocrine organs that release factors, termed adipokines, which affect other major organ systems. The development and functions of adipose tissues depend largely upon the glycosaminoglycan heparan sulfate. Heparan sulfate proteoglycans (HSPGs) surround both adipocytes and vascular structures and facilitate the communication between these two components. This communication mediates the continued export of adipokines from adipose tissues. Heparan sulfates regulate cellular physiology and communication through a sulfation code that ionically interacts with heparan-binding regions on a select set of proteins. Many of these proteins are growth factors and chemokines that regulate tissue function and inflammation. Cells regulate heparan sulfate sulfation through the release of heparanases and sulfatases. It is now possible to tissue engineer vascularized adipose tissues that express heparan sulfate proteoglycans. This makes it possible to use these tissue constructs to study the role of heparan sulfates in the regulation of adipokine production and release. It is possible to regulate the production of heparanases and sulfatases in order to fine-tune experimental studies.
Collapse
|
61
|
Richter HJ, Hauck AK, Batmanov K, Inoue SI, So BN, Kim M, Emmett MJ, Cohen RN, Lazar MA. Balanced control of thermogenesis by nuclear receptor corepressors in brown adipose tissue. Proc Natl Acad Sci U S A 2022; 119:e2205276119. [PMID: 35939699 PMCID: PMC9388101 DOI: 10.1073/pnas.2205276119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
Brown adipose tissue (BAT) is a key thermogenic organ whose expression of uncoupling protein 1 (UCP1) and ability to maintain body temperature in response to acute cold exposure require histone deacetylase 3 (HDAC3). HDAC3 exists in tight association with nuclear receptor corepressors (NCoRs) NCoR1 and NCoR2 (also known as silencing mediator of retinoid and thyroid receptors [SMRT]), but the functions of NCoR1/2 in BAT have not been established. Here we report that as expected, genetic loss of NCoR1/2 in BAT (NCoR1/2 BAT-dKO) leads to loss of HDAC3 activity. In addition, HDAC3 is no longer bound at its physiological genomic sites in the absence of NCoR1/2, leading to a shared deregulation of BAT lipid metabolism between NCoR1/2 BAT-dKO and HDAC3 BAT-KO mice. Despite these commonalities, loss of NCoR1/2 in BAT does not phenocopy the cold sensitivity observed in HDAC3 BAT-KO, nor does loss of either corepressor alone. Instead, BAT lacking NCoR1/2 is inflamed, particularly with respect to the interleukin-17 axis that increases thermogenic capacity by enhancing innervation. Integration of BAT RNA sequencing and chromatin immunoprecipitation sequencing data revealed that NCoR1/2 directly regulate Mmp9, which integrates extracellular matrix remodeling and inflammation. These findings reveal pleiotropic functions of the NCoR/HDAC3 corepressor complex in BAT, such that HDAC3-independent suppression of BAT inflammation counterbalances stimulation of HDAC3 activity in the control of thermogenesis.
Collapse
Affiliation(s)
- Hannah J. Richter
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Amy K. Hauck
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Kirill Batmanov
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Shin-Ichi Inoue
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Bethany N. So
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Mindy Kim
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Matthew J. Emmett
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Ronald N. Cohen
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL 60637
| | - Mitchell A. Lazar
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
62
|
Single-cell RNA sequencing of subcutaneous adipose tissues identifies therapeutic targets for cancer-associated lymphedema. Cell Discov 2022; 8:58. [PMID: 35725971 PMCID: PMC9209506 DOI: 10.1038/s41421-022-00402-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer-associated lymphedema frequently occurs following lymph node resection for cancer treatment. However, we still lack effective targeted medical therapies for the treatment or prevention of this complication. An in-depth elucidation of the cellular alterations in subcutaneous adipose tissues of lymphedema is essential for medical development. We performed single-cell RNA sequencing of 70,209 cells of the stromal vascular fraction of adipose tissues from lymphedema patients and healthy donors. Four subpopulations of adipose-derived stromal cells (ASCs) were identified. Among them, the PRG4+/CLEC3B+ ASC subpopulation c3 was significantly expanded in lymphedema and related to adipose tissue fibrosis. Knockdown of CLEC3B in vitro could significantly attenuate the fibrogenesis of ASCs from patients. Adipose tissues of lymphedema displayed a striking depletion of LYVE+ anti-inflammatory macrophages and exhibited a pro-inflammatory microenvironment. Pharmacological blockage of Trem1, an immune receptor predominantly expressed by the pro-inflammatory macrophages, using murine LR12, a dodecapeptide, could significantly alleviate lymphedema in a mouse tail model. Cell–cell communication analysis uncovered a perivascular ligand-receptor interaction module among ASCs, macrophages, and vascular endothelial cells. We provided a comprehensive analysis of the lineage–specific changes in the adipose tissues from lymphedema patients at a single-cell resolution. CLEC3B was found to be a potential target for alleviating adipose tissue fibrosis. Pharmacological blockage of TREM1 using LR12 could serve as a promising medical therapy for treating lymphedema.
Collapse
|
63
|
Rojas-Torres M, Sánchez-Gomar I, Rosal-Vela A, Beltrán-Camacho L, Eslava-Alcón S, Alonso-Piñeiro JÁ, Martín-Ramírez J, Moreno-Luna R, Durán-Ruiz MC. Assessment of endothelial colony forming cells delivery routes in a murine model of critical limb threatening ischemia using an optimized cell tracking approach. Stem Cell Res Ther 2022; 13:266. [PMID: 35729651 PMCID: PMC9210810 DOI: 10.1186/s13287-022-02943-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 01/15/2023] Open
Abstract
Background Endothelial colony forming cells (ECFCs), alone or in combination with mesenchymal stem cells, have been selected as potential therapeutic candidates for critical limb-threatening ischemia (CLTI), mainly for those patients considered as “no-option,” due to their capability to enhance revascularization and perfusion recovery of ischemic tissues. Nevertheless, prior to translating cell therapy to the clinic, biodistribution assays are required by regulatory guidelines to ensure biosafety as well as to discard undesired systemic translocations. Different approaches, from imaging technologies to qPCR-based methods, are currently applied. Methods In the current study, we have optimized a cell-tracking assay based on DiR fluorescent cell labeling and near-infrared detection for in vivo and ex vivo assays. Briefly, an improved protocol for DiR staining was set up, by incubation of ECFCs with 6.67 µM DiR and intensive washing steps prior cell administration. The minimal signal detected for the residual DiR, remaining after these washes, was considered as a baseline signal to estimate cell amounts correlated to the DiR intensity values registered in vivo. Besides, several assays were also performed to determine any potential effect of DiR over ECFCs functionality. Furthermore, the optimized protocol was applied in combination with qPCR amplification of specific human Alu sequences to assess the final distribution of ECFCs after intramuscular or intravenous administration to a murine model of CLTI. Results The optimized DiR labeling protocol indicated that ECFCs administered intramuscularly remained mainly within the hind limb muscle while cells injected intravenously were found in the spleen, liver and lungs. Conclusion Overall, the combination of DiR labeling and qPCR analysis in biodistribution assays constitutes a highly sensitive approach to systemically track cells in vivo. Thereby, human ECFCs administered intramuscularly to CLTI mice remained locally within the ischemic tissues, while intravenously injected cells were found in several organs. Our data corroborate the need to perform biodistribution assays in order to define specific parameters such as the optimal delivery route for ECFCs before their application into the clinic. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02943-8.
Collapse
Affiliation(s)
- Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Lucía Beltrán-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Sara Eslava-Alcón
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - José Ángel Alonso-Piñeiro
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | | | - Rafael Moreno-Luna
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain. .,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain.
| |
Collapse
|
64
|
Aratikatla A, Maffulli N, Rodriguez HC, Gupta M, Potty AG, El-Amin SF, Gupta A. Allogenic perinatal tissue for musculoskeletal regenerative medicine applications: a systematic review protocol. J Orthop Surg Res 2022; 17:307. [PMID: 35690774 PMCID: PMC9188718 DOI: 10.1186/s13018-022-03197-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Musculoskeletal ailments impact the lives of millions of people, and at times necessitate surgery followed by physiotherapy, drug treatments, or immobilization. Regenerative musculoskeletal medicine has undergone enormous progress over the last few decades. Sources of tissues used for regenerative medicine purposes can be grouped into autologous or allogenic. Although autologous sources are promising, there is a wide range of limitations with the treatment, including the lack of randomized controlled studies for orthopaedic conditions, donor site morbidity, and highly variable outcomes for patients. Allogenic sources bypass some of these shortcomings and are a promising source for orthopaedic regenerative medicine applications. METHODS A systematic search will be performed using PubMed, Elsevier, ScienceDirect, and Google Scholar databases for articles published in English before May 2022. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement and guidelines will be used. Studies will be eligible if they apply to acute and chronic orthopaedic musculoskeletal complications or animal or human disease models. Publications must include the use of MSCs and/or tissue obtained from amniotic/chorionic membrane, amniotic fluid, umbilical cord, and/or umbilical cord-derived Wharton's jelly as an intervention. Placebos, noninjury models, acute injury models, non-injury models, and gold standard treatments will be compared. The study selection will be performed by two independent reviewers using a dedicated reference management software. Data synthesis and meta-analysis will be performed separately for preclinical and clinical studies. DISCUSSION The results will be published in relevant peer-reviewed scientific journals. Investigators will present results at national or international conferences. TRIAL REGISTRATION The Protocol will be registered on PROSPERO international prospective register of systematic reviews prior to commencement.
Collapse
Affiliation(s)
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, 84084 Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Orthopedica” Department, Hospital of Salerno, 84124 Salerno, Italy
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4DG UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke-on-Trent, ST5 5BG UK
| | - Hugo C. Rodriguez
- Holy Cross Hospital, Orthopaedic Research Institute, Fort Lauderdale, FL 33334 USA
- Department of Orthopaedic Surgery, Larkin Community Hospital, South Miami, FL USA
| | - Manu Gupta
- Future Biologics, Lawrenceville, GA 30043 USA
- Polar Aesthetics Dental & Cosmetic Centre, Noida, Uttar Pradesh 201301 India
| | - Anish G. Potty
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
- Laredo Sports Medicine Clinic, Laredo, TX 78041 USA
| | - Saadiq F. El-Amin
- El-Amin Orthopaedic & Sports Medicine Institute, Lawrenceville, GA 30043 USA
- Regenerative Sports Medicine, Lawrenceville, GA 30043 USA
- BioIntegrate Inc., 2505 Newpoint Pkwy, Suite – 100, Lawrenceville, GA 30043 USA
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043 USA
- Polar Aesthetics Dental & Cosmetic Centre, Noida, Uttar Pradesh 201301 India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
- BioIntegrate Inc., 2505 Newpoint Pkwy, Suite – 100, Lawrenceville, GA 30043 USA
- Veterans in Pain (V.I.P.), Valencia, CA 91354 USA
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh 110048 India
| |
Collapse
|
65
|
Prišlin M, Vlahović D, Kostešić P, Ljolje I, Brnić D, Turk N, Lojkić I, Kunić V, Karadjole T, Krešić N. An Outstanding Role of Adipose Tissue in Canine Stem Cell Therapy. Animals (Basel) 2022; 12:ani12091088. [PMID: 35565514 PMCID: PMC9099541 DOI: 10.3390/ani12091088] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 01/15/2023] Open
Abstract
Adipose tissue, previously known as connective tissue with a role in energy storage, is currently changing the course of treatments in veterinary medicine. Recent studies have revealed one particularly impressive function among all the newly discovered functions of adipose tissue. The interactive cells hosted by adipose tissue, the stromal vascular fraction (SVF), and their role in treating numerous diseases have provided a prospective course of research with positive outcomes in regenerative veterinary medicine (RVM). This review describes the main features of adipose tissue, emphasizing an eclectic combination of cells within the SVF and its thus far researched therapeutic possibilities in canine RVM. An afterwards focus is on a highly researched component of the SVF, adipose-derived mesenchymal stem cells (ASCs), which were shown to have an extraordinary impact relying on several proposed mechanisms of action on mitigating pathologies in canines. Furthermore, ASC therapy showed the most significant results in the orthopaedics field and in neurology, dermatology, ophthalmology, gastroenterology, and hepatology, which elevates the possibilities of ASC therapy to a whole new level. Therefore, this review article aims to raise awareness of the importance of research on cellular components, within abundant and easily accessible adipose tissue, in the direction of regenerative therapy in canines, considering the positive outcomes so far. Although the focus is on the positive aspects of cellular therapy in canines, the researchers should not forget the importance of identifying the potential negative aspects within published and upcoming research. Safe and standardized treatment represents a fundamental prerequisite for positively impacting the lives of canine patients.
Collapse
Affiliation(s)
- Marina Prišlin
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (M.P.); (D.B.); (I.L.); (V.K.)
| | - Dunja Vlahović
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (D.V.); (P.K.); (N.T.); (T.K.)
| | - Petar Kostešić
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (D.V.); (P.K.); (N.T.); (T.K.)
| | - Ivana Ljolje
- Veterinary Clinic for Small Animals Buba, Dore Pfanove 11, 10000 Zagreb, Croatia;
| | - Dragan Brnić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (M.P.); (D.B.); (I.L.); (V.K.)
| | - Nenad Turk
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (D.V.); (P.K.); (N.T.); (T.K.)
| | - Ivana Lojkić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (M.P.); (D.B.); (I.L.); (V.K.)
| | - Valentina Kunić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (M.P.); (D.B.); (I.L.); (V.K.)
| | - Tugomir Karadjole
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (D.V.); (P.K.); (N.T.); (T.K.)
| | - Nina Krešić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia; (M.P.); (D.B.); (I.L.); (V.K.)
- Correspondence:
| |
Collapse
|
66
|
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
|
67
|
Hendawy H, Kaneda M, Yoshida T, Metwally E, Hambe L, Yoshida T, Shimada K, Tanaka R. Heterogeneity of Adipose Stromal Vascular Fraction Cells from the Different Harvesting Sites in Rats. Anat Rec (Hoboken) 2022; 305:3410-3421. [PMID: 35332993 DOI: 10.1002/ar.24915] [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: 02/01/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Abstract
In both veterinary and human health, regenerative medicine offers a promising cure for various disorders. One of the rate-limiting challenges in regenerative medicine is the considerable time and technique required to expand and grow cells in culture. Therefore, the stromal vascular fraction (SVF) shows a significant promise for various cell therapy approaches. The present study aimed to define and investigate the optimal harvest site of freshly isolated SVF cells from various adipose tissue (AT) depot sites in the female Sprague-Dawley (S.D.) rat. First, Hematoxylin and eosin (H&E) were used to analyze the morphological variations in AT samples from peri-ovarian, peri-renal, mesenteric, and omental sites. The presence of putative stromal cells positive CD34 was detected using immunohistochemistry. Then, the isolated SVF cells were examined for cell viability and cellular yield differences. Finally, the expression of mesenchymal stem cells and hematopoietic markers in the SVF cells subpopulation was studied using flow cytometry. The pluripotent gene expression profile was also evaluated. CD34 staining of the omental AT was substantially higher than those of other anatomical sites. Despite having the least quantity of fat, omental AT has the highest SVF cell fraction and viable cells. Along with CD90 and CD44 higher expression, Oct4, Sox2, and Rex-1 genes levels were higher in SVF cells isolated from the omental AT. To conclude, omental fat is the best candidate for SVF cell isolation in female S.D. rats with the highest SVF cell fraction with higher MSCs phenotypes and pluripotency gene expression.
Collapse
Affiliation(s)
- Hanan Hendawy
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo183-8509, Japan.,Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tadashi Yoshida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Elsayed Metwally
- Department of cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Egypt
| | - Lina Hambe
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo183-8509, Japan
| | - Tomohiko Yoshida
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo183-8509, Japan
| | - Kazumi Shimada
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo183-8509, Japan
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo183-8509, Japan
| |
Collapse
|
68
|
Xie H, Heier C, Meng X, Bakiri L, Pototschnig I, Tang Z, Schauer S, Baumgartner VJ, Grabner GF, Schabbauer G, Wolinski H, Robertson GR, Hoefler G, Zeng W, Wagner EF, Schweiger M, Zechner R. An immune-sympathetic neuron communication axis guides adipose tissue browning in cancer-associated cachexia. Proc Natl Acad Sci U S A 2022; 119:e2112840119. [PMID: 35210363 PMCID: PMC8892347 DOI: 10.1073/pnas.2112840119] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a hypermetabolic syndrome characterized by unintended weight loss due to the atrophy of adipose tissue and skeletal muscle. A phenotypic switch from white to beige adipocytes, a phenomenon called browning, accelerates CAC by increasing the dissipation of energy as heat. Addressing the mechanisms of white adipose tissue (WAT) browning in CAC, we now show that cachexigenic tumors activate type 2 immunity in cachectic WAT, generating a neuroprotective environment that increases peripheral sympathetic activity. Increased sympathetic activation, in turn, results in increased neuronal catecholamine synthesis and secretion, β-adrenergic activation of adipocytes, and induction of WAT browning. Two genetic mouse models validated this progression of events. 1) Interleukin-4 receptor deficiency impeded the alternative activation of macrophages, reduced sympathetic activity, and restrained WAT browning, and 2) reduced catecholamine synthesis in peripheral dopamine β-hydroxylase (DBH)-deficient mice prevented cancer-induced WAT browning and adipose atrophy. Targeting the intraadipose macrophage-sympathetic neuron cross-talk represents a promising therapeutic approach to ameliorate cachexia in cancer patients.
Collapse
Affiliation(s)
- Hao Xie
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Christoph Heier
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Xia Meng
- School of Medicine, Tsinghua University, 100190 Beijing, China
| | - Latifa Bakiri
- Genes and Disease Group, Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Zhiyuan Tang
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
- Department of Pharmacy, Affiliated Hospital of Nantong University, 226001 Nantong, China
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University Graz, 8010 Graz, Austria
| | | | - Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Gernot Schabbauer
- Institute of Physiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University Graz, 8010 Graz, Austria
| | - Wenwen Zeng
- School of Medicine, Tsinghua University, 100190 Beijing, China
| | - Erwin F Wagner
- Genes and Disease Group, Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Genes and Disease Group, Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria;
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria;
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
69
|
Luk C, Haywood NJ, Bridge KI, Kearney MT. Paracrine Role of the Endothelium in Metabolic Homeostasis in Health and Nutrient Excess. Front Cardiovasc Med 2022; 9:882923. [PMID: 35557517 PMCID: PMC9086712 DOI: 10.3389/fcvm.2022.882923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 02/02/2023] Open
Abstract
The vascular endothelium traditionally viewed as a simple physical barrier between the circulation and tissue is now well-established as a key organ mediating whole organism homeostasis by release of a portfolio of anti-inflammatory and pro-inflammatory vasoactive molecules. Healthy endothelium releases anti-inflammatory signaling molecules such as nitric oxide and prostacyclin; in contrast, diseased endothelium secretes pro-inflammatory signals such as reactive oxygen species, endothelin-1 and tumor necrosis factor-alpha (TNFα). Endothelial dysfunction, which has now been identified as a hallmark of different components of the cardiometabolic syndrome including obesity, type 2 diabetes and hypertension, initiates and drives the progression of tissue damage in these disorders. Recently it has become apparent that, in addition to vasoactive molecules, the vascular endothelium has the potential to secrete a diverse range of small molecules and proteins mediating metabolic processes in adipose tissue (AT), liver, skeletal muscle and the pancreas. AT plays a pivotal role in orchestrating whole-body energy homeostasis and AT dysfunction, characterized by local and systemic inflammation, is central to the metabolic complications of obesity. Thus, understanding and targeting the crosstalk between the endothelium and AT may generate novel therapeutic opportunities for the cardiometabolic syndrome. Here, we provide an overview of the role of the endothelial secretome in controlling the function of AT. The endothelial-derived metabolic regulatory factors are grouped and discussed based on their physical properties and their downstream signaling effects. In addition, we focus on the therapeutic potential of these regulatory factors in treating cardiometabolic syndrome, and discuss areas of future study of potential translatable and clinical significance. The vascular endothelium is emerging as an important paracrine/endocrine organ that secretes regulatory factors in response to nutritional and environmental cues. Endothelial dysfunction may result in imbalanced secretion of these regulatory factors and contribute to the progression of AT and whole body metabolic dysfunction. As the vascular endothelium is the first responder to local nutritional changes and adipocyte-derived signals, future work elucidating the changes in the endothelial secretome is crucial to improve our understanding of the pathophysiology of cardiometabolic disease, and in aiding our development of new therapeutic strategies to treat and prevent cardiometabolic syndrome.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
70
|
Park SS, Park M, Lee BT. Autologous stromal vascular fraction-loaded hyaluronic acid/gelatin-biphasic calcium phosphate scaffold for bone tissue regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 132:112533. [DOI: 10.1016/j.msec.2021.112533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022]
|
71
|
Slezak C, Rose R, Jilge JM, Nuster R, Hercher D, Slezak P. Physical Considerations for In Vitro ESWT Research Design. Int J Mol Sci 2021; 23:313. [PMID: 35008735 PMCID: PMC8745079 DOI: 10.3390/ijms23010313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/25/2021] [Indexed: 12/25/2022] Open
Abstract
In vitro investigations, which comprise the bulk of research efforts geared at identifying an underlying biomechanical mechanism for extracorporeal shock wave therapy (ESWT), are commonly hampered by inadequate descriptions of the underlying therapeutic acoustical pressure waves. We demonstrate the necessity of in-situ sound pressure measurements inside the treated samples considering the significant differences associated with available applicator technologies and cell containment. A statistical analysis of pulse-to-pulse variability in an electrohydraulic applicator yields a recommendation for a minimal pulse number of n = 300 for cell pallets and suspensions to achieve reproducible treatments. Non-linear absorption behavior of sample holders and boundary effects are shown for transient peak pressures and applied energies and may serve as a guide when in-situ measurements are not available or can be used as a controllable experimental design factor. For the use in microbiological investigations of ESWT we provide actionable identification of common problems in describing physical shockwave parameters and improving experimental setups by; (1) promoting in-situ sound field measurements, (2) statistical evaluation of applicator variability, and (3) extrapolation of treatment parameters based on focal and treatment volumes.
Collapse
Affiliation(s)
- Cyrill Slezak
- Department of Physics, Utah Valley University, Orem, UT 84059, USA;
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria; (R.R.); (J.M.J.); (D.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Roland Rose
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria; (R.R.); (J.M.J.); (D.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - Julia M. Jilge
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria; (R.R.); (J.M.J.); (D.H.)
- University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Robert Nuster
- Department of Physics, University of Graz, 8010 Graz, Austria;
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria; (R.R.); (J.M.J.); (D.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Paul Slezak
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria; (R.R.); (J.M.J.); (D.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
72
|
Caneparo C, Sorroza-Martinez L, Chabaud S, Fradette J, Bolduc S. Considerations for the clinical use of stem cells in genitourinary regenerative medicine. World J Stem Cells 2021; 13:1480-1512. [PMID: 34786154 PMCID: PMC8567446 DOI: 10.4252/wjsc.v13.i10.1480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The genitourinary tract can be affected by several pathologies which require repair or replacement to recover biological functions. Current therapeutic strategies are challenged by a growing shortage of adequate tissues. Therefore, new options must be considered for the treatment of patients, with the use of stem cells (SCs) being attractive. Two different strategies can be derived from stem cell use: Cell therapy and tissue therapy, mainly through tissue engineering. The recent advances using these approaches are described in this review, with a focus on stromal/mesenchymal cells found in adipose tissue. Indeed, the accessibility, high yield at harvest as well as anti-fibrotic, immunomodulatory and proangiogenic properties make adipose-derived stromal/SCs promising alternatives to the therapies currently offered to patients. Finally, an innovative technique allowing tissue reconstruction without exogenous material, the self-assembly approach, will be presented. Despite advances, more studies are needed to translate such approaches from the bench to clinics in urology. For the 21st century, cell and tissue therapies based on SCs are certainly the future of genitourinary regenerative medicine.
Collapse
Affiliation(s)
- Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Luis Sorroza-Martinez
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Julie Fradette
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| |
Collapse
|
73
|
Sharma S, Muthu S, Jeyaraman M, Ranjan R, Jha SK. Translational products of adipose tissue-derived mesenchymal stem cells: Bench to bedside applications. World J Stem Cells 2021; 13:1360-1381. [PMID: 34786149 PMCID: PMC8567449 DOI: 10.4252/wjsc.v13.i10.1360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
With developments in the field of tissue engineering and regenerative medicine, the use of biological products for the treatment of various disorders has come into the limelight among researchers and clinicians. Among all the available biological tissues, research and exploration of adipose tissue have become more robust. Adipose tissue engineering aims to develop by-products and their substitutes for their regenerative and immunomodulatory potential. The use of biodegradable scaffolds along with adipose tissue products has a major role in cellular growth, proliferation, and differentiation. Adipose tissue, apart from being the powerhouse of energy storage, also functions as the largest endocrine organ, with the release of various adipokines. The progenitor cells among the heterogeneous population in the adipose tissue are of paramount importance as they determine the capacity of regeneration of these tissues. The results of adipose-derived stem-cell assisted fat grafting to provide numerous growth factors and adipokines that improve vasculogenesis, fat graft integration, and survival within the recipient tissue and promote the regeneration of tissue are promising. Adipose tissue gives rise to various by-products upon processing. This article highlights the significance and the usage of various adipose tissue by-products, their individual characteristics, and their clinical applications.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
| | - Sathish Muthu
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul, Tamil Nadu 624304, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India.
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| |
Collapse
|
74
|
Efficacy and safety of autologous adipose-derived stromal vascular fraction enriched with platelet-rich plasma in flap repair of transsphincteric cryptoglandular fistulas. Tech Coloproctol 2021; 25:1301-1309. [PMID: 34606026 PMCID: PMC8580893 DOI: 10.1007/s10151-021-02524-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/07/2021] [Indexed: 12/26/2022]
Abstract
Background Transanal advancement flap repair of transsphincteric fistulas is a sphincter-preserving procedure, which frequently fails, probably due to ongoing inflammation in the remaining fistula tract. Adipose-derived stromal vascular fraction (SVF) has immunomodulatory properties promoting wound healing and suppressing inflammation. Platelet-rich plasma (PRP) reinforces this biological effect. The aim of this study was to evaluate the efficacy and safety of autologous adipose-derived SVF enriched with PRP in flap repair of transsphincteric cryptoglandular fistulas. Methods A prospective cohort study was conducted including consecutive patients with transsphincteric cryptoglandular fistula in a tertiary referral center. During flap repair, SVF was obtained by lipoharvesting and mechanical fractionation of adipose tissue and combined with PRP was injected around the internal opening and into the fistulous wall. Endpoints were fistula healing at clinical examination and fistula closure on postoperative magnetic resonance imaging (MRI). Adverse events were documented. Results Forty-five patients with transsphincteric cryptoglandular fistula were included (29 males, median age 44 years [range 36–53 years]). In the total study population, primary fistula healing was observed in 38 patients (84%). Among the 42 patients with intestinal continuity at time of surgery, primary fistula healing was observed in 35 patients (84%). In one patient, the fistula recurred, resulting in a long-term healing rate of 82%. MRI, performed in 37 patients, revealed complete closure of the fistula tract in 33 (89.2%). In the other patients, the tract was almost completely obliterated by scar tissue. During follow-up, none of these patients showed clinical signs of recurrence. The postoperative course was uneventful, except for three cases; venous thromboembolism in one patient and bleeding under the flap, necessitating intervention in two patients. Conclusions Addition of autologous SVF enriched with PRP during flap repair is feasible, safe and might improve outcomes in patients with a transsphincteric cryptoglandular fistula. Trial registration Dutch Trial Register, Trial Number: NL8416, https://www.trialregister.nl/
Collapse
|
75
|
Update on the Basic Science Concepts and Applications of Adipose-Derived Stem Cells in Hand and Craniofacial Surgery. Plast Reconstr Surg 2021; 148:475e-486e. [PMID: 34432707 DOI: 10.1097/prs.0000000000008279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
SUMMARY Adipose-derived stem cell therapy offers plastic surgeons a novel treatment alternative for conditions with few therapeutic options. Adipose-derived stem cells are a promising treatment because of their broad differentiation potential, capacity for self-renewal, and ease of isolation. Over the past decade, plastic surgeons have attempted to harness adipose-derived stem cells' unique cellular characteristics to improve the survival of traditional fat grafting procedures, a process known as cell-assisted lipotransfer. However, the full implications of cell-assisted lipotransfer in clinical practice remain incompletely understood, stressing the urgent need to assess the scientific evidence supporting adipose-derived stem cell-based interventions. Furthermore, with the strict regulatory climate surrounding tissue explantation therapies, reviewing the safety and efficacy of these treatments will clarify their regulatory viability moving forward. In this report, the authors provide a comprehensive, up-to-date appraisal of best evidence-based practices supporting adipose-derived stem cell-derived therapies, highlighting the known mechanisms behind current clinical applications in tissue engineering and regenerative medicine specific to plastic and reconstructive surgery. The authors outline best practices for the harvest and isolation of adipose-derived stem cells and discuss why procedure standardization will elucidate the scientific bases for their broad use. Finally, the authors discuss challenges posed by U.S. Food and Drug Administration oversight of these cell-based therapies and examine the role of adipose-derived stem cell-based applications in the future of plastic surgery.
Collapse
|
76
|
Laschke MW, Menger MD. The simpler, the better: tissue vascularization using the body's own resources. Trends Biotechnol 2021; 40:281-290. [PMID: 34404555 DOI: 10.1016/j.tibtech.2021.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
Tissue regeneration is crucially dependent on sufficient vascularization. In regenerative medicine, this can be effectively achieved by autologous vascularization strategies using the body's own resources. These strategies include the administration of blood-derived factor preparations, adipose tissue-based vascularization, and the in situ engineering of vascularized tissue. Due to their simplicity, the translation of these strategies into clinical practice is easier in terms of feasibility, safety requirements, and regulatory hurdles compared with complex and time-consuming procedures involving intensive cell manipulation. Hence, they are close to clinical application or are already being used to successfully treat patients by distinct personalized medicine concepts.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
77
|
Iannotta D, Yang M, Celia C, Di Marzio L, Wolfram J. Extracellular vesicle therapeutics from plasma and adipose tissue. NANO TODAY 2021; 39:101159. [PMID: 33968157 PMCID: PMC8104307 DOI: 10.1016/j.nantod.2021.101159] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Extracellular vesicles (EVs) are cell-released lipid-bilayer nanoparticles that contain biologically active cargo involved in physiological and pathological intercellular communication. In recent years, the therapeutic potential of EVs has been explored in various disease models. In particular, mesenchymal stromal cell-derived EVs have been shown to exert anti-inflammatory, anti-oxidant, anti-apoptotic, and pro-angiogenic properties in cardiovascular, metabolic and orthopedic conditions. However, a major drawback of EV-based therapeutics is scale-up issues due to extensive cell culture requirements and inefficient isolation protocols. An emerging alternative approach to time-consuming and costly cell culture expansion is to obtain therapeutic EVs directly from the body, for example, from plasma and adipose tissue. This review discusses isolation methods and therapeutic applications of plasma and adipose tissue-derived EVs, highlighting advantages and disadvantages compared to cell culture-derived ones.
Collapse
Affiliation(s)
- Dalila Iannotta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Man Yang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Christian Celia
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston TX, USA
| |
Collapse
|
78
|
Josh F, Soekamto TH, Adriani JR, Jonatan B, Mizuno H, Faruk M. The Combination of Stromal Vascular Fraction Cells and Platelet-Rich Plasma Reduces Malondialdehyde and Nitric Oxide Levels in Deep Dermal Burn Injury. J Inflamm Res 2021; 14:3049-3061. [PMID: 34267534 PMCID: PMC8275197 DOI: 10.2147/jir.s318055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/25/2021] [Indexed: 12/22/2022] Open
Abstract
Introduction Thermal burns release reactive oxygen species, which cause profound systemic and local changes. Stromal vascular fraction cells (SVFs) combined with platelet-rich plasma accelerate burn wound healing. This study investigated the effect of a combination of locally injected SVFs and PRP on malondialdehyde (MDA) and nitric oxide (NO) serum and tissue levels in a deep dermal burn model in Wistar rats. Methods Thirty-six adult Wistar rats weighing between 150 and 250 grams were used in this study to establish a deep dermal degree burn wound model. They were randomly divided into 4 groups: locally injected the combination SVFs and PRP, the Vaseline group, the placebo group, and healthy Wistar rats (the normal control group). MDA and NO levels in blood serum and burn wound tissue were measured at 8, 24, and 48 hours. Data were analyzed with one-way ANOVA followed by multiple comparisons tests and regression tests. Results Local injection of SVFs and PRP in combination affected blood MDA, tissue MDA, blood NO and tissue NO levels, with reductions of 0.257µmol/L, 0.427 µmol/L, 21.78nmol/mg, and 23.777nmol/mg, respectively. Injection of SVFs and PRP in combination reduced tissue MDA levels by 1.282 times, NO blood levels by 2.305, and NO tissue levels by 2.377 times compared to Vaseline application. Conclusion The combination of SVFs and PRP undeniably reduced the MDA and NO levels in blood and tissue compared to those in the Vaseline and placebo groups. The injection of these two preparations in combination inhibited the local and systemic stress oxidative response, as illustrated by the decreased MDA and NO levels in blood serum and tissue.
Collapse
Affiliation(s)
- Fonny Josh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia.,Department of Plastic and Reconstructive Surgery, Wahidin Sudirohusodo Hospital, Makassar, South Sulawesi, Indonesia
| | | | - Januar Rizky Adriani
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| | - Billy Jonatan
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University, Tokyo, Japan
| | - Muhammad Faruk
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| |
Collapse
|
79
|
Josh F, Soekamto TH, Adriani JR, Jonatan B, Mizuno H, Faruk M. The Combination of Stromal Vascular Fraction Cells and Platelet-Rich Plasma Reduces Malondialdehyde and Nitric Oxide Levels in Deep Dermal Burn Injury. J Inflamm Res 2021; Volume 14:3049-3061. [DOI: https:/doi.org/10.2147/jir.s318055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
80
|
Cellular and physiological circadian mechanisms drive diurnal cell proliferation and expansion of white adipose tissue. Nat Commun 2021; 12:3482. [PMID: 34108488 PMCID: PMC8190103 DOI: 10.1038/s41467-021-23770-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 04/29/2021] [Indexed: 02/05/2023] Open
Abstract
Hyperplastic expansion of white adipose tissue (WAT) relies in part on the proliferation of adipocyte precursor cells residing in the stromal vascular cell fraction (SVF) of WAT. This study reveals a circadian clock- and feeding-induced diurnal pattern of cell proliferation in the SVF of visceral and subcutaneous WAT in vivo, with higher proliferation of visceral adipocyte progenitor cells subsequent to feeding in lean mice. Fasting or loss of rhythmic feeding eliminates this diurnal proliferation, while high fat feeding or genetic disruption of the molecular circadian clock modifies the temporal expression of proliferation genes and impinges on diurnal SVF proliferation in eWAT. Surprisingly, high fat diet reversal, sufficient to reverse elevated SVF proliferation in eWAT, was insufficient in restoring diurnal patterns of SVF proliferation, suggesting that high fat diet induces a sustained disruption of the adipose circadian clock. In conclusion, the circadian clock and feeding simultaneously impart dynamic, regulatory control of adipocyte progenitor proliferation, which may be a critical determinant of adipose tissue expansion and health over time.
Collapse
|
81
|
A Novel Hypothesis and Characterization to Isolate Microvascular Endothelial Cells Simultaneously with Adipose-Derived Stem Cells from the Human Adipose-Derived Stromal Vascular Fraction. Tissue Eng Regen Med 2021; 18:429-440. [PMID: 33877617 DOI: 10.1007/s13770-021-00332-5] [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: 12/22/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Angiogenesis and vasculogenesis are essential processes for successful tissue regeneration in tissue engineering and regenerative medicine. The adipose-derived stromal vascular fraction (SVF) is not only a source of adipose stem cells (ASC) but also a suitable source of microvascular endothelial cells because it is a rich capillary network. So, we propose a new hypothesis for isolating adipose-derived human microvascular endothelial cells (HMVEC-A) from the SVF and developed a dual isolation system that isolates two cell types from one tissue. METHOD To isolate HMVEC-A, we analyzed the supernatant discarded when ASC is isolated from the adipose-derived SVF. Based on this analysis, we assumed that the SVF adherent to the bottom of the culture plate was divided into two fractions: the stromal fraction as the ASC-rich fraction, and the vascular fraction (VF) as the endothelial cells-rich fraction floating in the culture supernatant. VF isolation was optimized and the efficiency was compared, and the endothelial cells characteristics of HMVEC-A were confirmed by flow cytometric analysis, immunocytochemistry (ICC), a DiI-acetylated low-density lipoprotein (DiI-Ac-LDL) uptake, and in vitro tube formation assay. RESULTS Consistent with the hypothesis, we found a large population of HMVEC-A in the VF and isolated these HMVEC-A by our isolation method. Additionally, this method had higher yields and shorter doubling times than other endothelial cells isolation methods and showed typical morphological and phenotypic characteristics of endothelial cells. CONCLUSION Cells obtained by the method according to our hypothesis can be applied as a useful source for studies such as tissue-to-tissue networks, angiogenesis and tissue regeneration, patient-specific cell therapy, and organoid chips.
Collapse
|
82
|
Valizadeh A, Asghari S, Bastani S, Sarvari R, Keyhanvar N, Razin SJ, Khiabani AY, Yousefi B, Yousefi M, Shoae-Hassani A, Mahmoodpoor A, Hamishehkar H, Tavakol S, Keshel SH, Nouri M, Seifalian AM, Keyhanvar P. Will stem cells from fat and growth factors from blood bring new hope to female patients with reproductive disorders? Reprod Biol 2021; 21:100472. [PMID: 33639342 DOI: 10.1016/j.repbio.2020.100472] [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: 08/22/2020] [Revised: 11/21/2020] [Accepted: 12/06/2020] [Indexed: 01/05/2023]
Abstract
Female reproductive system disorders (FRSD) with or without infertility are prevalent women's health problems with a variety of treatment approaches including surgery and hormone therapy. It currently considering to sub-branch of regenerative medicine including stem cells or growth factors injection-based delivery treatment might be improved female reproductive health life. The most common products used for these patients treatment are autologous cell or platelet-based products from patients, including platelet-rich plasma, plasma rich in growth factor, platelet-rich fibrin, and stromal vascular fraction. In this review, we discuss each of the above products used in treatment of FRSD and critically evaluate the clinical outcome.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cell Research Center, Stem Cells and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Asghari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Bastani
- Stem Cell Research Center, Stem Cells and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raana Sarvari
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Keyhanvar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Gene Yakhteh Keyhan (Genik) Company (Ltd), Pharmaceutical Biotechnology Incubator, Tabriz University of Medical Sciences, Tabriz, Iran; ARTAN1100 Startup Accelerator, Tabriz, Iran
| | - Sepideh Jalilzadeh Razin
- Stem Cell Research Center, Stem Cells and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Yousefzadeh Khiabani
- Department of Epidemiology and Reproductive Health, Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Shoae-Hassani
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Andam Baft Yakhteh (ABY) Company (Ltd), Tehran, Iran
| | - Ata Mahmoodpoor
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Heidari Keshel
- Medical Nanotechnology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Stem Cells and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; ARTAN1100 Startup Accelerator, Tabriz, Iran; Zist Andam Yakhteh Azerbaijan (ZAYA) Company (PHT), Medical Instrument Technology Incubator, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alexander Marcus Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre (Ltd), The London Innovation Bio Science Centre, London NW1 0NH, United Kingdom
| | - Peyman Keyhanvar
- Stem Cell Research Center, Stem Cells and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; ARTAN1100 Startup Accelerator, Tabriz, Iran; Zist Andam Yakhteh Azerbaijan (ZAYA) Company (PHT), Medical Instrument Technology Incubator, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; HealthNBICS Group, Convergence of Knowledge and Technology to the benefit of Society Network (CKTSN), Universal Scientific Education and Research Network (USERN), Tabriz, Iran.
| |
Collapse
|
83
|
Stromal-vascular fraction of adipose tissue as an alternative source of cellular material for regenerative medicine. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.01.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue is the most convenient source of cellular material for regenerative medicine as it can be obtained in significant quantities via cosmetic liposuction, lipoaspiration of subcutaneous fat or by excision of fat deposits. Adipose tissue consists of adipocytes and cells, which are the part of the stromal-vascular fraction (SVF). Different cell populations can be isolated from SVF, among which the population of adipose tissue stem cells (adipose-derived stem cells, ADSC) is especially important for regenerative medicine. SVF can be obtained relatively easily from adipose tissue (adipose tissue is an alternative to bone marrow in terms of being a source of stem cells) and used to treat various pathologies. Recent studies show that SVF not only has a therapeutic effect similar to that of ADSC, but in some cases is even more effective. The article provides the analysis of the main methods of SVF obtainment, characteristics of SVF cellular composition, its potential for use in clinical medicine and its main advantages over other sources of cellular material, including ADSC cultured in vitro, for regenerative medicine. Keywords: adipocytes, adipose-derived stem cells, regenerative medicine, stromal-vascular fraction
Collapse
|
84
|
Wan CR, Muya L, Kansara V, Ciulla TA. Suprachoroidal Delivery of Small Molecules, Nanoparticles, Gene and Cell Therapies for Ocular Diseases. Pharmaceutics 2021; 13:pharmaceutics13020288. [PMID: 33671815 PMCID: PMC7926337 DOI: 10.3390/pharmaceutics13020288] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Suprachoroidal drug delivery technology has advanced rapidly and emerged as a promising administration route for a variety of therapeutic candidates, in order to target multiple ocular diseases, ranging from neovascular age-related macular degeneration to choroidal melanoma. This review summarizes the latest preclinical and clinical progress in suprachoroidal delivery of therapeutic agents, including small molecule suspensions, polymeric entrapped small molecules, gene therapy (viral and nonviral nanoparticles), viral nanoparticle conjugates (VNCs), and cell therapy. Formulation customization is critical in achieving favorable pharmacokinetics, and sustained drug release profiles have been repeatedly observed for multiple small molecule suspensions and polymeric formulations. Novel therapeutic agents such as viral and nonviral gene therapy, as well as VNCs, have demonstrated promise in animal studies. Several of these suprachoroidally-administered therapies have been assessed in clinical trials, including small molecule suspensions of triamcinolone acetonide and axitinib, viral vector RGX-314 for gene therapy, and VNC AU-011. With continued drug delivery research and optimization, coupled with customized drug formulations, suprachoroidal drug delivery may address large unmet therapeutic needs in ophthalmology, targeting affected tissues with novel therapies for efficacy benefits, compartmentalizing therapies away from unaffected tissues for safety benefits, and achieving durability to relieve the treatment burden noted with current agents.
Collapse
|
85
|
Nie F, Bi H, Zhang C, Ding P. Differentiation potential and mRNA profiles of human dedifferentiated adipose cells and adipose‑derived stem cells from young donors. Mol Med Rep 2020; 23:47. [PMID: 33200799 PMCID: PMC7705993 DOI: 10.3892/mmr.2020.11685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Dedifferentiated adipose cells (DAs) and adipose-derived stem cells (ADSCs) are two of the primary types of stem cells derived from adipose tissue, which have been reported to possess similar characteristics, but also exhibit unique phenotypic and functional advantages. However, several reports have described inconsistent results regarding their differences in multilineage differentiation function. Moreover, to the best of our knowledge, there are no studies assessing their myogenic ability, or the differences in the transcriptome between the two cell types derived from lipoaspirates via tumescent liposuction from the same donors. The aim of the present study was to compare the properties and expression profiles of these cell types. Subcutaneous adipose tissue of three female patients (aged 23–30 years) with a physiological BMI (19.1–23.9 kg/m2) were obtained during tumescent liposuction of the abdomen or the thigh. The stromal vascular fraction and mature adipocytes were obtained via collagenase digestion, and ADSCs and DAs were cultured successively. To determine the differences between DAs and ADSCs after 6–7 passages, cell proliferation assays, phenotypic assessment, differentiation assays and high-throughput RNA sequencing (seq) were used. Similar cell morphologies, proliferation dynamics, surface markers and transcriptome expression profiles were observed between the DAs and ADSCs. Whilst there were notable individual differences in the osteogenic, lipogenic, chondrogenic and myogenic abilities of the DAs and ADSCs, it was difficult to determine their differentiation potential based only on the cell source. Interestingly, the myogenic ability was relatively stronger in cells with relatively weaker lipogenic ability. Only 186 differentially expressed genes between the two groups were identified using RNAseq. Several of these genes were involved in biological functions such as transcription regulation, protein translation regulation, cytokine interactions and energy metabolism regulation. The results of the present study suggested a similar functional potential of DAs and ADSCs from young donors undergoing tumescent liposuction operation in regeneration areas and the balance of the differentiative ability of the same cell populations. These data may provide a foundation for further clinical administration of stem cells derived from adipose tissues in therapy.
Collapse
Affiliation(s)
- Fangfei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Chen Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Pengbing Ding
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
86
|
Vidal L, Brennan MÁ, Krissian S, De Lima J, Hoornaert A, Rosset P, Fellah BH, Layrolle P. In situ production of pre-vascularized synthetic bone grafts for regenerating critical-sized defects in rabbits. Acta Biomater 2020; 114:384-394. [PMID: 32688088 DOI: 10.1016/j.actbio.2020.07.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/16/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
Abstract
Reconstructing large bone defects caused by severe trauma or resection of tumors remains a challenge for surgeons. A fibula free flap and its vascularized bed can be transplanted to the reconstruction site to achieve healing. However, this technique adds morbidity, and requires microsurgery and sculpting of the bone tissue to adapt the graft to both the vasculature and the anatomy of the defect. The aim of the current study was to evaluate an alternative approach consisting of the in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a critical-sized bone defect. 3D printed chambers containing biphasic calcium phosphate (BCP) granules, perfused by a local vascular pedicle, with or without the addition of stromal vascular fraction (SVF), were subcutaneously implanted into New Zealand White female rabbits. SVF was prepared extemporaneously from autologous adipose tissue, the vascular pedicle was isolated from the inguinal site, while BCP granules alone served as a control group. After 8 weeks, the constructs containing a vascular pedicle exhibited abundant neovascularization with blood vessels sprouting from the pedicle, leading to significantly increased vascularization compared to BCP controls. Pre-vascularized synthetic bone grafts were then transplanted into 15 mm critical-sized segmental ulnar defects for a further 8 weeks. Micro-CT and decalcified histology revealed that pre-vascularization of synthetic bone grafts led to enhanced bone regeneration. This pre-clinical study demonstrates the feasibility and efficacy of the in situ production of pre-vascularized synthetic bone grafts for regenerating large bone defects, thereby addressing an important clinical need. STATEMENT OF SIGNIFICANCE: The current gold standard in large bone defect regeneration is vascularized fibula grafting. An alternative approach consisting of in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a bone defect is presented here. 3D printed chambers were filled with biphasic calcium phosphate granules, supplemented with autologous stromal vascular fraction and an axial vascular pedicle and subcutaneously implanted in inguinal sites. These pre-vascularized synthetic grafts were then transplanted into critical-sized segmental ulnar defects. Micro-CT and decalcified histology revealed that the pre-vascularized synthetic bone grafts led to higher bone regeneration than non-vascularized constructs. An alternative to vascularized fibula grafting is provided and may address an important clinical need for large bone defect reconstruction.
Collapse
|
87
|
Couselo‐Seijas M, Lopez‐Canoa JN, Fernandez ÁL, González‐Melchor L, Seoane LM, Duran‐Muñoz D, Rozados‐Luis A, González‐Juanatey JR, Rodríguez‐Mañero M, Eiras S. Inflammatory and lipid regulation by cholinergic activity in epicardial stromal cells from patients who underwent open-heart surgery. J Cell Mol Med 2020; 24:10958-10969. [PMID: 32767737 PMCID: PMC7521153 DOI: 10.1111/jcmm.15727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/26/2020] [Accepted: 07/18/2020] [Indexed: 01/14/2023] Open
Abstract
The modulation of acetylcholine (ACh) release by botulinum toxin injection into epicardial fat diminishes atrial fibrillation (AF) recurrence. These results suggest an interaction between autonomic imbalance and epicardial fat as risk factors of AF. Our aim was to study the inflammatory, lipidic and fibroblastic profile of epicardial stroma from patients who underwent open-heart surgery, their regulation by cholinergic activity and its association with AF. We performed in vitro and ex vivo assays from paired subcutaneous and epicardial stromal cells or explants from 33 patients. Acute ACh effects in inflammation and lipid-related genes were analysed by qPCR, in intracellular calcium mobilization were performed by Fluo-4 AM staining and in neutrophil migration by trans-well assays. Chronic ACh effects on lipid accumulation were visualized by AdipoRed. Plasma protein regulation by parasympathetic denervation was studied in vagotomized rats. Our results showed a higher pro-inflammatory profile in epicardial regarding subcutaneous stromal cells. Acute ACh treatment up-regulated monocyte chemoattractant protein 1 levels. Chronic ACh treatment improved lipid accumulation within epicardial stromal cells (60.50% [22.82-85.13] vs 13.85% [6.17-23.16], P < .001). Additionally, patients with AF had higher levels of fatty acid-binding protein 4 (1.54 ± 0.01 vs 1.47 ± 0.01, P = .005). Its plasma levels were pronouncedly declined in vagotomized rats (2.02 ± 0.21 ng/mL vs 0.65 ± 0.23 ng/mL, P < .001). Our findings support the characterization of acute or chronic cholinergic activity on epicardial stroma and its association with AF.
Collapse
Affiliation(s)
- Marinela Couselo‐Seijas
- Translational Cardiology groupHealth Research InstituteSantiago de CompostelaSpain
- University of Santiago de CompostelaSantiago de CompostelaSpain
| | - José N. Lopez‐Canoa
- Translational Cardiology groupHealth Research InstituteSantiago de CompostelaSpain
- Cardiovascular DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
| | - Ángel L. Fernandez
- University of Santiago de CompostelaSantiago de CompostelaSpain
- Heart Surgery DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- CIBERCVMadridSpain
| | | | - Luisa M. Seoane
- Endocrine Physiopathology GroupHealth Research InstituteSantiago de CompostelaSpain
- CIBERobnMadridSpain
| | - Darío Duran‐Muñoz
- Heart Surgery DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- CIBERCVMadridSpain
| | - Adriana Rozados‐Luis
- Translational Cardiology groupHealth Research InstituteSantiago de CompostelaSpain
| | - José Ramón González‐Juanatey
- University of Santiago de CompostelaSantiago de CompostelaSpain
- Cardiovascular DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- Cardiology GroupHealth Research InstituteSantiago de CompostelaSpain
- CIBERCVMadridSpain
| | - Moisés Rodríguez‐Mañero
- Translational Cardiology groupHealth Research InstituteSantiago de CompostelaSpain
- Cardiovascular DepartmentUniversity Hospital of Santiago de CompostelaSantiago de CompostelaSpain
- CIBERCVMadridSpain
| | - Sonia Eiras
- Translational Cardiology groupHealth Research InstituteSantiago de CompostelaSpain
- CIBERCVMadridSpain
| |
Collapse
|
88
|
Groppa E, Colliva A, Vuerich R, Kocijan T, Zacchigna S. Immune Cell Therapies to Improve Regeneration and Revascularization of Non-Healing Wounds. Int J Mol Sci 2020; 21:E5235. [PMID: 32718071 PMCID: PMC7432547 DOI: 10.3390/ijms21155235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
With the increased prevalence of chronic diseases, non-healing wounds place a significant burden on the health system and the quality of life of affected patients. Non-healing wounds are full-thickness skin lesions that persist for months or years. While several factors contribute to their pathogenesis, all non-healing wounds consistently demonstrate inadequate vascularization, resulting in the poor supply of oxygen, nutrients, and growth factors at the level of the lesion. Most existing therapies rely on the use of dermal substitutes, which help the re-epithelialization of the lesion by mimicking a pro-regenerative extracellular matrix. However, in most patients, this approach is not efficient, as non-healing wounds principally affect individuals afflicted with vascular disorders, such as peripheral artery disease and/or diabetes. Over the last 25 years, innovative therapies have been proposed with the aim of fostering the regenerative potential of multiple immune cell types. This can be achieved by promoting cell mobilization into the circulation, their recruitment to the wound site, modulation of their local activity, or their direct injection into the wound. In this review, we summarize preclinical and clinical studies that have explored the potential of various populations of immune cells to promote skin regeneration in non-healing wounds and critically discuss the current limitations that prevent the adoption of these therapies in the clinics.
Collapse
Affiliation(s)
- Elena Groppa
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (E.G.); (A.C.); (R.V.); (T.K.)
| | - Andrea Colliva
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (E.G.); (A.C.); (R.V.); (T.K.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Roman Vuerich
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (E.G.); (A.C.); (R.V.); (T.K.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Tea Kocijan
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (E.G.); (A.C.); (R.V.); (T.K.)
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (E.G.); (A.C.); (R.V.); (T.K.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
89
|
Svalgaard JD, Juul S, Vester-Glovinski PV, Haastrup EK, Ballesteros OR, Lynggaard CD, Jensen AK, Fischer-Nielsen A, Herly M, Munthe-Fog L. Lipoaspirate Storage Time and Temperature: Effects on Stromal Vascular Fraction Quality and Cell Composition. Cells Tissues Organs 2020; 209:54-63. [PMID: 32580198 DOI: 10.1159/000507825] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/10/2020] [Indexed: 11/19/2022] Open
Abstract
The adipose tissue-derived stromal vascular fraction (SVF) is a promising candidate for use in cell therapy and tissue engineering due to its regenerative and immunomodulatory properties. Some therapies are based on using the complete SVF product, whereas others depend on the expansion of adipose-derived stromal cells (ASCs) in culture. The latter application often involves a time delay between adipose tissue harvest and SVF isolation. This study investigated how storage time and temperature affected cell quality and composition. Aliquots of lipoaspirate were stored cold (4°C), at room temperature (18-20°C), or at 37°C. SVF was isolated on sequential time points over a period of 48 h, and the following were assessed: cell viability, vitality, composition, and the proliferative potential of the ASCs. When the lipoaspirate was stored cold, the viability of the SVF remained stable for up to 48 h; however, the vitality of the SVF decreased significantly after 24 h. When stored at higher temperatures (room temperature or 37°C), the vitality of the SVF decreased after 8 h. The ASC fraction in the SVF decreased rapidly after 8 h when stored at higher temperatures, whereas this change was delayed significantly when the lipoaspirate was stored cold. Tendencies towards increases in the lag phase, population doubling time (PDt), and time to reach confluency were observed when the lipoaspirate was stored at higher temperatures. The vitality of the SVF was correlated significantly with the time of the lag phase and the time required to reach confluence, whereas no correlation was observed with the PDt. Both prolonged storage time and increased temperature during lipoaspirate storage negatively affected the quality of the obtained SVF. Our results suggest that lipoaspirate should be stored for no longer than 24 h at 4°C to maintain the optimal quality for the isolation of SVF and the expansion of ASCs.
Collapse
Affiliation(s)
| | - Sarah Juul
- Department of Plastic Surgery and Burns, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Eva Kannik Haastrup
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Olga Rivera Ballesteros
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Duch Lynggaard
- Department of Otorhinolaryngology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Anne Fischer-Nielsen
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Herly
- Department of Plastic Surgery and Burns, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lea Munthe-Fog
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
90
|
Sun Y, Chen S, Zhang X, Pei M. Significance of Cellular Cross-Talk in Stromal Vascular Fraction of Adipose Tissue in Neovascularization. Arterioscler Thromb Vasc Biol 2020; 39:1034-1044. [PMID: 31018663 DOI: 10.1161/atvbaha.119.312425] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adult stem cell-based therapy has been regarded as a promising treatment for tissue ischemia because of its ability to promote new blood vessel formation. Bone marrow-derived mesenchymal stem cells are the most used angiogenic cells for therapeutic neovascularization, yet the side effects and low efficacy have limited their clinical application. Adipose stromal vascular fraction is an easily accessible, heterogeneous cell system comprised of endothelial, stromal, and hematopoietic cell lineages, which has been shown to spontaneously form robust, patent, and functional vasculatures in vivo. However, the characteristics of each cell population and their specific roles in neovascularization remain an area of ongoing investigation. In this review, we summarize the functional capabilities of various stromal vascular fraction constituents during the process of neovascularization and attempt to analyze whether the cross-talk between these constituents generates a synergetic effect, thus contributing to the development of new potential therapeutic strategies to promote neovascularization.
Collapse
Affiliation(s)
- Yuan Sun
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Song Chen
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Xicheng Zhang
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Ming Pei
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| |
Collapse
|
91
|
Xue P, Hou Y, Zuo Z, Wang Z, Ren S, Dong J, Fu J, Wang H, Andersen ME, Zhang Q, Xu Y, Pi J. Long isoforms of NRF1 negatively regulate adipogenesis via suppression of PPARγ expression. Redox Biol 2019; 30:101414. [PMID: 31931283 PMCID: PMC6957832 DOI: 10.1016/j.redox.2019.101414] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/02/2019] [Accepted: 12/23/2019] [Indexed: 01/12/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 1 (NRF1), a ubiquitously expressed CNC-bZIP transcription factor, plays a critical role in white adipocyte (WAC) biology, whereas the underlying mechanisms remain unknown. The mouse Nrf1 gene is transcribed in a number of alternatively spliced forms, resulting in two long protein isoforms (L-NRF1) containing 741 and 742 amino acids (aa) and multiple short isoforms (S-NRF1). Our previous study found that adipocyte-specific knockout of Nrf1 [Nrf1(f)-KO] in mice disturbs the expression of lipolytic genes in adipocytes, leading to adipocyte hypertrophy followed by inflammation, pyroptosis and insulin resistance. In the present study, we found that the stromal vascular fraction (SVF) cells isolated from white adipose tissues (WAT) of Nrf1(f)-KO mice display augmented adipogenesis showing elevated mRNA and protein expression of adipogenic markers and lipid accumulation. In 3T3-L1 cells, stable knockdown (KD) of all or long isoforms of Nrf1 (termed as A-Nrf1-KD and L-Nrf1-KD, respectively) using lentiviral shRNAs resulted in enhanced and accelerated adipogenic differentiation. Conversely, overexpression of L-NRF1-741, but not any of the S-NRF1, substantially attenuated adipogenesis in 3T3-L1 cells. These findings indicate that L-NRF1 might serve as a critical negative regulator of adipogenesis. Mechanistic investigation revealed that L-NRF1 may negatively regulates the transcription of peroxisome proliferator-activated receptor γ (PPARγ), in particular the master regulator of adipogenesis PPARγ2. Taken all together, the findings in the present study provide further evidence for a novel role of NRF1 beyond its participation in cellular antioxidant response and suggest that L-NRF1 is a negative regulator of PPARγ2 expression and thereby can suppress adipogenesis. SVF cells isolated from WAT of Nrf1(f)-KO mice displayed augmented adipogenesis. Stable silencing of L-Nrf1 in 3T3-L1 cells resulted in enhanced and accelerated adipogenesis. Overexpression of L-NRF1-741, but not S-NRF1s, attenuated adipogenesis in 3T3-L1 cells. L-NRF1 suppressed adipogenesis via downregulating PPARγ2 expression.
Collapse
Affiliation(s)
- Peng Xue
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; ScitoVation LLC, Research Triangle Park, NC, USA
| | - Yongyong Hou
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Zhuo Zuo
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Zhendi Wang
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Suping Ren
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Jian Dong
- ScitoVation LLC, Research Triangle Park, NC, USA
| | - Jingqi Fu
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Huihui Wang
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | | | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yuanyuan Xu
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China.
| | - Jingbo Pi
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
92
|
Trivisonno A, Alexander RW, Baldari S, Cohen SR, Di Rocco G, Gentile P, Magalon G, Magalon J, Miller RB, Womack H, Toietta G. Intraoperative Strategies for Minimal Manipulation of Autologous Adipose Tissue for Cell- and Tissue-Based Therapies: Concise Review. Stem Cells Transl Med 2019; 8:1265-1271. [PMID: 31599497 PMCID: PMC6877766 DOI: 10.1002/sctm.19-0166] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/10/2019] [Indexed: 12/16/2022] Open
Abstract
The stromal vascular fraction (SVF) is a heterogeneous population of stem/stromal cells isolated from perivascular and extracellular matrix (ECM) of adipose tissue complex (ATC). Administration of SVF holds a strong therapeutic potential for regenerative and wound healing medicine applications aimed at functional restoration of tissues damaged by injuries or chronic diseases. SVF is commonly divided into cellular stromal vascular fraction (cSVF) and tissue stromal vascular fraction (tSVF). Cellular SVF is obtained from ATC by collagenase digestion, incubation/isolation, and pelletized by centrifugation. Enzymatic disaggregation may alter the relevant biological characteristics of adipose tissue, while providing release of complex, multiattachment of cell-to-cell and cell-to-matrix, effectively eliminating the bioactive ECM and periadventitial attachments. In many countries, the isolation of cellular elements is considered as a "more than minimal" manipulation, and is most often limited to controlled clinical trials and subject to regulatory review. Several alternative, nonenzymatic methods of adipose tissue processing have been developed to obtain via minimal mechanical manipulation an autologous tSVF product intended for delivery, reducing the procedure duration, lowering production costs, decreasing regulatory burden, and shortening the translation into the clinical setting. Ideally, these procedures might allow for the integration of harvesting and processing of adipose tissue for ease of injection, in a single procedure utilizing a nonexpanded cellular product at the point of care, while permitting intraoperative autologous cellular and tissue-based therapies. Here, we review and discuss the options, advantages, and limitations of the major strategies alternative to enzymatic processing currently developed for minimal manipulation of adipose tissue. Stem Cells Translational Medicine 2019;8:1265&1271.
Collapse
Affiliation(s)
- Angelo Trivisonno
- Department of Surgical ScienceUniversity of Rome “La Sapienza”RomeItaly
| | | | - Silvia Baldari
- Department of ResearchAdvanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer InstituteRomeItaly
- Department of Medical Surgical Sciences and BiotechnologiesUniversity of Rome “La Sapienza”LatinaItaly
| | - Steven R. Cohen
- FACES+ Plastic SurgerySkin and Laser Center and the University of CaliforniaSan DiegoCaliforniaUSA
| | - Giuliana Di Rocco
- Department of ResearchAdvanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Pietro Gentile
- Department of Plastic and Reconstructive SurgeryUniversity of Rome Tor VergataRomeItaly
| | - Guy Magalon
- Plastic Surgery DepartmentAssistance Publique Hôpitaux de Marseille (APHM), Aix Marseille UniversityMarseilleFrance
| | - Jérémy Magalon
- Vascular Research Center of MarseilleAix Marseille University, INSERM UMR 1076MarseilleFrance
- Cell Therapy LaboratoryCBT‐1409, INSERM, Assistance Publique Hôpitaux de MarseilleMarseilleFrance
| | | | - Hayley Womack
- FACES+ Plastic SurgerySkin and Laser Center and the University of CaliforniaSan DiegoCaliforniaUSA
| | - Gabriele Toietta
- Department of ResearchAdvanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
93
|
Bashir MM, Sohail M, Ahmad FJ, Choudhery MS. Preenrichment with Adipose Tissue-Derived Stem Cells Improves Fat Graft Retention in Patients with Contour Deformities of the Face. Stem Cells Int 2019; 2019:5146594. [PMID: 31827528 PMCID: PMC6886337 DOI: 10.1155/2019/5146594] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/08/2019] [Accepted: 08/30/2019] [Indexed: 02/05/2023] Open
Abstract
Quick absorption of adipose tissue grafts makes the outcomes less satisfactory for clinical applications. In the current study, adipose tissue grafts were mixed with adipose tissue-derived stem cells (ASCs) to improve retention of adipose tissue grafts and to make the clinical outcomes of fat grafting more reliable. Adipose tissue was either injected alone (conventional group) or mixed with ASCs (stem cell group) before injection. In both groups, adipose tissue was injected at the site of contour throughout layers of tissues till visual clinical symmetry with the opposite side was achieved. The volume of injected fat graft was measured after 72 hours and 6 months using a B-mode ultrasound device connected with a 12 MH frequency probe. The percentage reduction in the volume of injected fat, physician satisfaction scores (Ph-SCs), and patient satisfaction scores (P-SCs) were also recorded. After 6 months, there was significantly lower fat absorption in the stem cell group as compared to the conventional group. Mean physician and patient satisfaction scores were significantly improved in the stem cell group. No significant adverse effects were noted in any patient. Significantly lower absorption of graft due to the use of ASCs improves the clinical outcomes of conventional fat grafting for contour deformities of the face. The current preenrichment strategy is noninvasive, safe and can be applied to other diseases that require major tissue augmentation such as breast surgery. This trial is registered with NCT02494752.
Collapse
Affiliation(s)
- Muhammad M. Bashir
- Plastic Surgery Department, King Edward Medical University/Mayo Hospital, Lahore, Pakistan
| | - Muhammad Sohail
- Plastic Surgery Department, King Edward Medical University/Mayo Hospital, Lahore, Pakistan
| | - Fridoon J. Ahmad
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
| | - Mahmood S. Choudhery
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
| |
Collapse
|
94
|
Bauknight DK, Osinski V, Dasa SSK, Nguyen AT, Marshall MA, Hartman J, Harms M, O’Mahony G, Boucher J, Klibanov AL, McNamara CA, Kelly KA. Importance of thorough tissue and cellular level characterization of targeted drugs in the evaluation of pharmacodynamic effects. PLoS One 2019; 14:e0224917. [PMID: 31725756 PMCID: PMC6855449 DOI: 10.1371/journal.pone.0224917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022] Open
Abstract
Targeted nanoparticle delivery is a promising strategy for increasing efficacy and limiting side effects of therapeutics. When designing a targeted liposomal formulation, the in vivo biodistribution of the particles must be characterized to determine the value of the targeting approach. Peroxisome proliferator-activated receptor (PPAR) agonists effectively treat metabolic syndrome by decreasing dyslipidemia and insulin resistance but side effects have limited their use, making them a class of compounds that could benefit from targeted liposomal delivery. The adipose targeting sequence peptide (ATS) could fit this role, as it has been shown to bind to adipose tissue endothelium and induce weight loss when delivered conjugated to a pro-apoptotic peptide. To date, however, a full assessment of ATS in vivo biodistribution has not been reported, leaving important unanswered questions regarding the exact mechanisms whereby ATS targeting enhances therapeutic efficacy. We designed this study to evaluate the biodistribution of ATS-conjugated liposomes loaded with the PPARα/γ dual agonist tesaglitazar in leptin-deficient ob/ob mice. The ATS-liposome biodistribution in adipose tissue and other organs was examined at the cellular and tissue level using microscopy, flow cytometry, and fluorescent molecular tomography. Changes in metabolic parameters and gene expression were measured by target and off-target tissue responses to the treatment. Unexpectedly, ATS targeting did not increase liposomal uptake in adipose relative to other tissues, but did increase uptake in the kidneys. Targeting also did not significantly alter metabolic parameters. Analysis of the liposome cellular distribution in the stromal vascular fraction with flow cytometry revealed high uptake by multiple cell types. Our findings highlight the need for thorough study of in vivo biodistribution when evaluating a targeted therapy.
Collapse
Affiliation(s)
- Dustin K. Bauknight
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
- Cancer Center, University of Virginia, Charlottesville, VA, United States of America
| | - Victoria Osinski
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
| | - Siva Sai Krishna Dasa
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
- Cancer Center, University of Virginia, Charlottesville, VA, United States of America
| | - Anh T. Nguyen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - Melissa A. Marshall
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - Julia Hartman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - Matthew Harms
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gavin O’Mahony
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jeremie Boucher
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Alexander L. Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - Kimberly A. Kelly
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
- Cancer Center, University of Virginia, Charlottesville, VA, United States of America
- * E-mail:
| |
Collapse
|
95
|
The Effect of Early Rounds of ex vivo Expansion and Cryopreservation on the Adipogenic Differentiation Capacity of Adipose-Derived Stromal/Stem Cells. Sci Rep 2019; 9:15943. [PMID: 31685852 PMCID: PMC6828715 DOI: 10.1038/s41598-019-52086-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022] Open
Abstract
Multipotent adipose-derived stromal/stem cells (ASCs) are candidates for use in cellular therapies for the treatment of a variety of conditions/diseases. Ex vivo expansion of freshly isolated ASCs may be necessary prior to clinical application to ensure that clinically relevant cell numbers are administered during treatment. In addition, cryopreserving cells at early passages allows for storage of freshly isolated cells for extended periods of time before expanding these cells for clinical usage. There are however several concerns that these laboratory-based procedures may alter the characteristics of the cells and in so doing decrease their regenerative potential. In this study we report on the impact of early rounds of cryopreservation (P0) and ex vivo expansion (P0 to P5) on the phenotypic characteristics and adipogenic differentiation potential of ASCs. Our results show that ASCs that upregulate CD36 expression during adipogenic differentiation gradually decrease with increasing expansion rounds. The consequent decrease in adipogenic differentiation capacity was evident in both gene expression and flow cytometry-based phenotypic studies. Successive rounds of expansion did not however alter cell surface marker expression of the cells. We also show that early cryopreservation of ASCs (at P0) does not affect the adipogenic differentiation potential of the cells.
Collapse
|
96
|
Stefanis AJ, Groh T, Arenbergerova M, Arenberger P, Bauer PO. Stromal Vascular Fraction and its Role in the Management of Alopecia: A Review. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2019; 12:35-44. [PMID: 32038756 PMCID: PMC6937163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Adipose cells organized in small clusters under the reticular dermis closely interact with hair follicular cells and regulate the hair cycle. Intradermal adipocyte progenitor cells are activated toward the end of the telogen phase to proliferate and differentiate into mature adipocytes. These cells, surrounding the hair follicles, secrete signaling molecules that control the progression of the hair cycle. Diseases associated with defects in adipocyte homeostasis, such as lipodystrophy and focal dermal hypoplasia, lead to alopecia. In this review, we discuss the potential influence of stromal vascular fraction from adipose tissue in the management of alopecia as well as its involvement in preclinical and clinical trials.
Collapse
Affiliation(s)
- Athanasios J Stefanis
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Tomas Groh
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Monika Arenbergerova
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Petr Arenberger
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Peter O Bauer
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| |
Collapse
|
97
|
Montgomery MK, Bayliss J, Keenan S, Rhost S, Ting SB, Watt MJ. The role of Ap2a2 in PPARα-mediated regulation of lipolysis in adipose tissue. FASEB J 2019; 33:13267-13279. [PMID: 31533003 DOI: 10.1096/fj.201900909rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adipose tissue plays a major role in the regulation of systemic metabolic homeostasis, with the AP2 adaptor complex being important in clathrin-mediated endocytosis (CME) of various cell surface receptors, including glucose transporter 4, the insulin receptor, and β-adrenergic receptors (ARs). One of the AP2 subunits, adaptor-related protein complex 2, α2 subunit (Ap2a2), has recently been identified as a peroxisome proliferator-activated receptor (PPAR)α target gene. The effects of PPARα on the AP2 adaptor complex and CME are unknown. We generated adipocyte-specific Ap2a2 knockout mice and investigated their metabolism when fed a standard chow or high-fat diet, without and with supplementation with the PPARα-agonist WY-14643 (WY). Although Ap2a2 deletion had only minor effects on glycaemic control, it led to substantial impairment in β-adrenergic activation of lipolysis, as evidenced by a loss of cAMP response, PKA activation, and glycerol/fatty acid release. These differences were related to increased cell surface localization of the β2- and β3-ARs. Lipolytic defects were accompanied by impaired WY-mediated loss of fat mass and whole-body fat oxidation. This study demonstrates a novel role for PPARα in β-adrenergic regulation of adipose tissue lipolysis and for adipose tissue in supplying adequate substrate to other peripheral tissues to accommodate the increase in systemic fatty acid oxidation that occurs upon treatment with PPARα agonists.-Montgomery, M. K., Bayliss, J., Keenan, S., Rhost, S., Ting, S. B., Watt, M. J. The role of Ap2a2 in PPARα-mediated regulation of lipolysis in adipose tissue.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Jacqueline Bayliss
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Stacey Keenan
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Sarah Rhost
- Australian Centre for Blood Diseases, The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | - Stephen B Ting
- Australian Centre for Blood Diseases, The Alfred Centre, Monash University, Melbourne, Victoria, Australia.,Department of Haematology, Box Hill Hospital (Eastern Health), Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
98
|
Ferdowsi Khosroshahi A, Soleimani Rad J, Kheirjou R, Roshangar B, Rashtbar M, Salehi R, Ranjkesh MR, Roshangar L. Adipose tissue‐derived stem cells upon decellularized ovine small intestine submucosa for tissue regeneration: An optimization and comparison method. J Cell Physiol 2019; 235:1556-1567. [DOI: 10.1002/jcp.29074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Ahad Ferdowsi Khosroshahi
- Department of Anatomical Sciences, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Jafar Soleimani Rad
- Department of Anatomical Sciences, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Razie Kheirjou
- Department of Anatomical Sciences, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Babak Roshangar
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Morteza Rashtbar
- Stem Cell and Regenerative Medicine Research Center Guilan University of Medical Science Rasht Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Biomedical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Reza Ranjkesh
- Department of Dermatology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Leila Roshangar
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
99
|
Ayyappan JP, Ganapathi U, Lizardo K, Vinnard C, Subbian S, Perlin DS, Nagajyothi JF. Adipose Tissue Regulates Pulmonary Pathology during TB Infection. mBio 2019; 10:e02771-18. [PMID: 30992360 PMCID: PMC6469978 DOI: 10.1128/mbio.02771-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis infection, remains a major cause of mortality and morbidity worldwide. One-third of the world population is infected with M. tuberculosis, and about 15 million people with latent tuberculosis infection (LTBI) reside in the United States. An estimated 10% of individuals with LTBI are at risk of progressing to active disease. Loss of body mass, or wasting, accompanied by a significant reduction of body fat is often associated with active TB disease and is considered to be immunosuppressive and a major determinant of severity and outcome of disease. While the lungs are the primary site of M. tuberculosis infection and TB manifestation, recent reports have shown that adipose tissue serves as an important reservoir for M. tuberculosis In this article, we investigated the association between M. tuberculosis infection, adipose tissue, and TB disease progression using a transgenic inducible "fatless" model system, the FAT-ATTAC (fat apoptosis through targeted activation of caspase 8) mouse. By selectively ablating fat tissue during M. tuberculosis infection, we directly tested the role of fat cell loss and adipose tissue physiology in regulating pulmonary pathology, bacterial burden, and immune status. Our results confirm the presence of M. tuberculosis in fat tissue after aerosol infection of mice and show that loss of fat cells is associated with an increase in pulmonary M. tuberculosis burden and pathology. We conclude that acute loss of adipose tissue during LTBI may predispose the host to active TB disease.IMPORTANCE Although the lungs are the port of entry and the predominant site of TB disease manifestation, we and others have demonstrated that M. tuberculosis also persists in adipose tissue of aerosol-infected animals and directly or indirectly alters adipose tissue physiology, which in turn alters whole-body immuno-metabolic homeostasis. Our present report demonstrates a direct effect of loss of adipocytes (fat cells) on promoting the severity of pulmonary pathogenesis during TB, advancing our understanding of the pathogenic interactions between wasting and TB activation/reactivation.
Collapse
Affiliation(s)
- Janeesh Plakkal Ayyappan
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Usha Ganapathi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Kezia Lizardo
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Christopher Vinnard
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - David S. Perlin
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Jyothi F Nagajyothi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| |
Collapse
|
100
|
Magalon J, Velier M, Simoncini S, François P, Bertrand B, Daumas A, Benyamine A, Boissier R, Arnaud L, Lyonnet L, Fernandez S, Dignat-George F, Casanova D, Guillet B, Granel B, Paul P, Sabatier F. Molecular profile and proangiogenic activity of the adipose-derived stromal vascular fraction used as an autologous innovative medicinal product in patients with systemic sclerosis. Ann Rheum Dis 2019; 78:391-398. [PMID: 30612118 DOI: 10.1136/annrheumdis-2018-214218] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The autologous stromal vascular fraction (SVF) from adipose tissue is an alternative to cultured adipose-derived stem cells for use in regenerative medicine and represents a promising therapy for vasculopathy and hand disability in systemic sclerosis (SSc). However, the bioactivity of autologous SVF is not documented in this disease context. This study aimed to compare the molecular and functional profiles of the SVF-based medicinal product obtained from SSc and healthy subjects. METHODS Good manufacturing practice (GMP)-grade SVF from 24 patients with SSc and 12 healthy donors (HD) was analysed by flow cytometry to compare the distribution of the CD45- and CD45+ haematopoietic cell subsets. The ability of SVF to form a vascular network was assessed using Matrigel in vivo assay. The transcriptomic and secretory profiles of the SSc-SVF were assessed by RNA sequencing and multiplex analysis, respectively, and were compared with the HD-SVF. RESULTS The distribution of the leucocyte, endothelial, stromal, pericyte and transitional cell subsets was similar for SSc-SVF and HD-SVF. SSc-SVF retained its vasculogenic capacity, but the density of neovessels formed in SVF-loaded Matrigel implanted in nude mice was slightly decreased compared with HD-SVF. SSc-SVF displayed a differential molecular signature reflecting deregulation of angiogenesis, endothelial activation and fibrosis. CONCLUSIONS Our study provides the first evidence that SSc does not compromise the vascular repair capacity of SVF, supporting its use as an innovative autologous biotherapy. The characterisation of the specific SSc-SVF molecular profile provides new perspectives for delineating markers of the potency of SVF and its targets for the treatment of SSc.
Collapse
Affiliation(s)
- Jérémy Magalon
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Mélanie Velier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | | | - Pauline François
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Baptiste Bertrand
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Aurélie Daumas
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Audrey Benyamine
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Romain Boissier
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Urology Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Laurent Arnaud
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Luc Lyonnet
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | | | - Françoise Dignat-George
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Dominique Casanova
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Benjamin Guillet
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- CERIMED, Aix-Marseille University, AP-HM, Marseille, France
| | - Brigitte Granel
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Pascale Paul
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| |
Collapse
|