1
|
Gandolfi S, Sanouj A, Chaput B, Coste A, Sallerin B, Varin A. The role of adipose tissue-derived stromal cells, macrophages and bioscaffolds in cutaneous wound repair. Biol Direct 2024; 19:85. [PMID: 39343924 PMCID: PMC11439310 DOI: 10.1186/s13062-024-00534-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Skin healing is a complex and dynamic physiological process that follows mechanical alteration of the skin barrier. Under normal conditions, this complex process can be divided into at least three continuous and overlapping phases: an inflammatory reaction, a proliferative phase that leads to tissue reconstruction and a phase of tissue remodeling. Macrophages critically contribute to the physiological cascade for tissue repair. In fact, as the inflammatory phase progresses, macrophage gene expression gradually shifts from pro-inflammatory M1-like to pro-resolutive M2-like characteristics, which is critical for entry into the repair phase. A dysregulation in this macrophage' shift phenotype leads to the persistence of the inflammatory phase. Mesenchymal stromal cells and specifically the MSC-derived from adipose tissue (ADSCs) are more and more use to treat inflammatory diseases and several studies have demonstrated that ADSCs promote the wound healing thanks to their neoangiogenic, immunomodulant and regenerative properties. In several studies, ADSCs and macrophages have been injected directly into the wound bed, but the delivery of exogenous cells directly to the wound raise the problem of cell engraftment and preservation of pro-resolutive phenotype and viability of the cells. Complementary approaches have therefore been explored, such as the use of biomaterials enriched with therapeutic cell to improve cell survival and function. This review will present a background of the current scaffold models, using adipose derived stromal-cells and macrophage as therapeutic cells for wound healing, through a discussion on the potential impact for future applications in skin regeneration. According to the PRISMA statement, we resumed data from investigations reporting the use ADSCs and bioscaffolds and data from macrophages behavior with functional biomaterials in wound healing models. In the era of tissue engineering, functional biomaterials, that can maintain cell delivery and cellular viability, have had a profound impact on the development of dressings for the treatment of chronic wounds. Promising results have been showed in pre-clinical reports using ADSCs- and macrophages-based scaffolds to accelerate and to improve the quality of the cutaneous healing.
Collapse
Affiliation(s)
- S Gandolfi
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France.
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 1 Av. Pr.Jean Poulhès, 31400, Toulouse, France.
| | - A Sanouj
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| | - B Chaput
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 1 Av. Pr.Jean Poulhès, 31400, Toulouse, France
| | - A Coste
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| | - B Sallerin
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
- Department of Pharmacology, Toulouse University Hospital, 1 Av Pr.Jean Poulhès, 31400, Toulouse, France
| | - A Varin
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| |
Collapse
|
2
|
Hernández-Melchor D, Ortiz G, Madrazo I, Suarez JJ, Barrera N, Porchia LM, Chávez F, Velázquez-Aranda F, González-Espinosa A, Padilla A, López-Bayghen E. Improvement of endometrial thickness and in vitro fertilization outcomes in patients with Asherman's refractory endometrium using autologous mesenchymal stem cells from the stromal vascular fraction. Am J Transl Res 2024; 16:4020-4031. [PMID: 39262711 PMCID: PMC11384418 DOI: 10.62347/uagf1249] [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: 02/23/2024] [Accepted: 07/02/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE Patients with Asherman's Syndrome (AS) and an endometrial thickness (EMT) less than 7 mm are infertile women with suboptimal endometrium due to uterine scarring or endometrial atrophy. This study aimed to examine the effect of intrauterine injections of adipose-derived mesenchymal stem cells (ADMSC) from the Stromal Vascular Fraction (SVF) of adipose tissue on EMT and in vitro fertilization (IVF) outcomes: which are improvements in EMT and pregnancy rates. METHODS This double-arm retrospective study included 41 AS patients with hysteroscopic adhesiolysis. Twenty-one patients with AS refractory endometrium (Group 2) were given ADMSC to improve EMT, and 20 non-treated, age-matched patients served as controls (Group 1). For Group 2, SVF was isolated from 15 ml of adipose tissue and transmyometrial injected into the patient's uterine cavity. For all patients, EMT was examined using ultrasound before embryo transfer. RESULTS In Group 2, after ADMSC treatment, EMT significantly improved (3.2 ± 1.8 mm, P<0.001). Afterward, three patients spontaneously became pregnant, and eighteen underwent frozen embryo transfer. A significant increase in implantation (66.7% vs. 4.8%, P = 0.002) and live birth rates (0.0% vs. 47.6%, P = 0.001) were recorded. No significant difference was observed in EMT, cycle implantation, or clinical pregnancy between the two groups, but the live birth rate in Group 2 after ADMSC treatment was higher than in Group 1. CONCLUSION The results demonstrate that autologous intrauterine ADMSC injection can improve EMT, implantation, and pregnancy rates in AS patients with refractory endometrium. This research underscores the life-changing potential of autologous ADMSC treatment for patients with refractory endometrium, providing a promising avenue for future treatments.
Collapse
Affiliation(s)
- Dinorah Hernández-Melchor
- Science, Technology and Society Program, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, México
- Instituto Regenera SC, México City, México
| | - Ginna Ortiz
- Investigación Clínica, Instituto de Infertilidad y Genética Ingenes México, México City, México
| | - Iván Madrazo
- Investigación Clínica, Instituto de Infertilidad y Genética Ingenes México, México City, México
| | - Juan José Suarez
- Investigación Clínica, Instituto de Infertilidad y Genética Ingenes México, México City, México
| | - Norma Barrera
- Instituto de Fertilidad y Genética Puebla Puebla, México
| | | | | | | | | | - América Padilla
- Science, Technology and Society Program, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, México
| | - Esther López-Bayghen
- Science, Technology and Society Program, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, México
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN) México City, México
| |
Collapse
|
3
|
Dhoundiyal S, Alam MA. Advancements in Biotechnology and Stem Cell Therapies for Breast Cancer Patients. Curr Stem Cell Res Ther 2024; 19:1072-1083. [PMID: 37815191 DOI: 10.2174/011574888x268109230924233850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
This comprehensive review article examines the integration of biotechnology and stem cell therapy in breast cancer diagnosis and treatment. It discusses the use of biotechnological tools such as liquid biopsies, genomic profiling, and imaging technologies for accurate diagnosis and monitoring of treatment response. Stem cell-based approaches, their role in modeling breast cancer progression, and their potential for breast reconstruction post-mastectomy are explored. The review highlights the importance of personalized treatment strategies that combine biotechnological tools and stem cell therapies. Ethical considerations, challenges in clinical translation, and regulatory frameworks are also addressed. The article concludes by emphasizing the potential of integrating biotechnology and stem cell therapy to improve breast cancer outcomes, highlighting the need for continued research and collaboration in this field.
Collapse
Affiliation(s)
- Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| |
Collapse
|
4
|
Umer A, Ghouri MD, Muyizere T, Aqib RM, Muhaymin A, Cai R, Chen C. Engineered Nano-Bio Interfaces for Stem Cell Therapy. PRECISION CHEMISTRY 2023; 1:341-356. [PMID: 37654807 PMCID: PMC10466455 DOI: 10.1021/prechem.3c00056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 09/02/2023]
Abstract
Engineered nanomaterials (ENMs) with different topographies provide effective nano-bio interfaces for controlling the differentiation of stem cells. The interaction of stem cells with nanoscale topographies and chemical cues in their microenvironment at the nano-bio interface can guide their fate. The use of nanotopographical cues, in particular nanorods, nanopillars, nanogrooves, nanofibers, and nanopits, as well as biochemical forces mediated factors, including growth factors, cytokines, and extracellular matrix proteins, can significantly impact stem cell differentiation. These factors were seen as very effective in determining the proliferation and spreading of stem cells. The specific outgrowth of stem cells can be decided with size variation of topographic nanomaterial along with variation in matrix stiffness and surface structure like a special arrangement. The precision chemistry enabled controlled design, synthesis, and chemical composition of ENMs can regulate stem cell behaviors. The parameters of size such as aspect ratio, diameter, and pore size of nanotopographic structures are the main factors for specific termination of stem cells. Protein corona nanoparticles (NPs) have shown a powerful facet in stem cell therapy, where combining specific proteins could facilitate a certain stem cell differentiation and cellular proliferation. Nano-bio reactions implicate the interaction between biological entities and nanoparticles, which can be used to tailor the stem cells' culmination. The ion release can also be a parameter to enhance cellular proliferation and to commit the early differentiation of stem cells. Further research is needed to fully understand the mechanisms underlying the interactions between engineered nano-bio interfaces and stem cells and to develop optimized regenerative medicine and tissue engineering designs.
Collapse
Affiliation(s)
- Arsalan Umer
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
| | - Muhammad Daniyal Ghouri
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
| | - Theoneste Muyizere
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Raja Muhammad Aqib
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Abdul Muhaymin
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Rong Cai
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
- GBA
National Institute for Nanotechnology Innovation, Guangdong 5110700, China
| |
Collapse
|
5
|
Hernández-Melchor D, Palafox-Gómez C, Madrazo I, Ortiz G, Padilla-Viveros A, López-Bayghen E. Surgical and nutritional interventions for endometrial receptivity: A case report and review of literature. World J Clin Cases 2022; 10:12295-12304. [PMID: 36483831 PMCID: PMC9724549 DOI: 10.12998/wjcc.v10.i33.12295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/16/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is an endocrine disease that combines metabolic, reproductive, and psychological dysfunctions. Ovulation disorders and impaired endometrial receptivity in PCOS can cause infertility. Insulin resistance (IR) is a pathological state of inadequate response to insulin that affects reproduction in PCOS, as damage caused by IR at the endometrial level becomes an obstacle for embryo implantation. Reversing IR resulted in spontaneous pregnancies in PCOS patients, indicating that metabolic corrections improve endometrial dysfunctions. Mesenchymal stem-cell treatment has also corrected endometrial quality and lead to pregnancies in patients with Asherman’s syndrome. We propose a combination of nutritional intervention with the surgical placement of stem cells to improve endometrial quality to achieve pregnancy in a PCOS patient undergoing in vitro fertilization (IVF) treatment.
CASE SUMMARY After two failed IVF cycles, a metabolic intervention, consisting of a ketogenic diet with daily consumption of 50 g of carbohydrates (CH), was indicated until pregnancy. Metabolic Syndrome was assessed using the Harmonizing Definition (3 of 5 pathologies: Central obesity, hypertension, hyperglycemia, hypertriglyceridemia, and dyslipidemia), and the Homeostatic Model Assessment of IR (HOMA-IR) was used to measure the level of IR. Once IR improved, endometrial quality improved. However, two day 5-thawed embryos (euploid, donated oocyte–partner's sperm) failed to implant, suggesting endometrial quality improvement was insufficient. Therefore, transmyometrial implantation of mesenchymal stem cells from the stromal vascular fraction of adipose tissue was performed to enrich the endometrial stem cell niche. Minimal endometrial mean thickness for embryo transfer (6.9 mm) was achieved three months after stem cell treatment and continuous dietary control of IR. Two euploid-day 5-thawed embryos (donated oocyte–partner's sperm) were transferred, and embryo implantation was confirmed on day 14 by β-hCG serum levels. Currently, a 37 wk baby girl is born.
CONCLUSION In PCOS, endometrial quality can be improved by combining nutrient-based metabolic correction with endometrial stem cell niche enrichment.
Collapse
Affiliation(s)
- Dinorah Hernández-Melchor
- Science, Technology and Society Program, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City 07360, CDMX, México
- Clinical Research, Instituto Regenera SC, México City 05320, CDMX, México
| | - Cecilia Palafox-Gómez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City 07360, CDMX, México
| | - Ivan Madrazo
- Investigación Clínica, Instituto de Infertilidad y Genética México SC, INGENES, México City 05320, CDMX, México
| | - Ginna Ortiz
- Investigación Clínica, Instituto de Infertilidad y Genética México SC, INGENES, México City 05320, CDMX, México
| | - America Padilla-Viveros
- Science, Technology and Society Program, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City 07360, CDMX, México
| | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, CDMX, México
| |
Collapse
|
6
|
PLGA/Gelatin/Hyaluronic Acid Fibrous Membrane Scaffold for Therapeutic Delivery of Adipose-Derived Stem Cells to Promote Wound Healing. Biomedicines 2022; 10:biomedicines10112902. [PMID: 36428471 PMCID: PMC9687264 DOI: 10.3390/biomedicines10112902] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Hyaluronic acid (HA) has been suggested to be a preferential material for the delivery of adipose-derived stem cells (ASCs) in wound healing. By incorporating HA in electrospun poly (lactide-co-glycolide) (PLGA)/gelatin (PG) fibrous membrane scaffolds (FMS), we aim to fabricate PLGA/gelatin/HA (PGH) FMS to provide a milieu for 3D culture and delivery of ASCs. The prepared FMS shows adequate cytocompatibility and is suitable for attachment and growth of ASCs. Compared with PG, the PGH offers an enhanced proliferation rate of ASCs, shows higher cell viability, and better maintains an ASC-like phenotype during in vitro cell culture. The ASCs in PGH also show upregulated expression of genes associated with angiogenesis and wound healing. From a rat full-thickness wound healing model, a wound treated with PGH/ASCs can accelerate the wound closure rate compared with wounds treated with PGH, alginate wound dressing, and gauze. From H&E and Masson's trichrome staining, the PGH/ASC treatment can promote wound healing by increasing the epithelialization rate and forming well-organized dermis. This is supported by immunohistochemical staining of macrophages and α-smooth muscle actin, where early recruitment of macrophages, macrophage polarization, and angiogenesis was found due to the delivered ASCs. The content of type III collagen is also higher than type I collagen within the newly formed skin tissue, implying scarless wound healing. Taken together, using PGH FMS as a topical wound dressing material for the therapeutic delivery of ASCs, a wound treated with PGH/ASCs was shown to accelerate wound healing significantly in rats, through modulating immunoreaction, promoting angiogenesis, and reducing scar formation at the wound sites.
Collapse
|
7
|
Romaniyanto FNU, Mahyudin F, Prakoeswa CRS, Notobroto HB, Tinduh D, Ausrin R, Rantam FA, Suroto H, Utomo DN, Rhatomy S. Adipose-Derived Stem Cells (ASCs) for Regeneration of Intervertebral Disc Degeneration: Review Article. STEM CELLS AND CLONING: ADVANCES AND APPLICATIONS 2022; 15:67-76. [DOI: 10.2147/sccaa.s379714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/08/2022] [Indexed: 11/05/2022]
|
8
|
Kim YH, Im GB, Kim SW, Kim YJ, Yu T, Lee JR, Um SH, Joung YK, Bhang SH. Anti-senescence ion-delivering nanocarrier for recovering therapeutic properties of long-term-cultured human adipose-derived stem cells. J Nanobiotechnology 2021; 19:352. [PMID: 34717632 PMCID: PMC8557526 DOI: 10.1186/s12951-021-01098-7] [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: 07/16/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Human adipose-derived stem cells (hADSCs) have been used in various fields of tissue engineering because of their promising therapeutic efficacy. However, the stemness of hADSCs cannot be maintained for long durations, and their therapeutic cellular functions, such as paracrine factor secretion decrease during long-term cell culture. To facilitate the use of long-term-cultured hADSCs (L-ADSCs), we designed a novel therapeutic anti-senescence ion-delivering nanocarrier (AIN) that is capable of recovering the therapeutic properties of L-ADSCs. In the present study, we introduced a low-pH-responsive ion nanocarrier capable of delivering transition metal ions that can enhance angiogenic paracrine factor secretion from L-ADSCs. The AINs were delivered to L-ADSCs in an intracellular manner through endocytosis. RESULTS Low pH conditions within the endosomes induced the release of transition metal ions (Fe) into the L-ADSCs that in turn caused a mild elevation in the levels of reactive oxygen species (ROS). This mild elevation in ROS levels induced a downregulation of senescence-related gene expression and an upregulation of stemness-related gene expression. The angiogenic paracrine factor secretion from L-ADSCs was significantly enhanced, and this was evidenced by the observed therapeutic efficacy in response to treatment of a wound-closing mouse model with conditioned medium obtained from AIN-treated L-ADSCs that was similar to that observed in response to treatment with short-term-cultured adipose-derived stem cells. CONCLUSIONS This study suggests a novel method and strategy for cell-based tissue regeneration that can overcome the limitations of the low stemness and therapeutic efficacy of stem cells that occurs during long-term cell culture.
Collapse
Affiliation(s)
- Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Taekyung Yu
- Department of Chemical Engineering, College of Engineering, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Ju-Ro Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seoungbuk-gu, Seoul, 02792, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seoungbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, 113 Gwahangno, Yuseong-gu, Daejeon, 305-333, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea.
| |
Collapse
|
9
|
Tatsis D, Vasalou V, Kotidis E, Anestiadou E, Grivas I, Cheva A, Koliakos G, Venetis G, Pramateftakis MG, Ouzounidis N, Angelopoulos S. The Combined Use of Platelet-Rich Plasma and Adipose-Derived Mesenchymal Stem Cells Promotes Healing. A Review of Experimental Models and Future Perspectives. Biomolecules 2021; 11:biom11101403. [PMID: 34680036 PMCID: PMC8533225 DOI: 10.3390/biom11101403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
Wound healing and tissue regeneration are a field of clinical medicine presenting high research interest, since various local and systematic factors can inhibit these processes and lead to an inferior result. New methods of healing enhancement constantly arise, which, however, require experimental validation before their establishment in everyday practice. Platelet-rich plasma (PRP) is a well-known autologous factor that promotes tissue healing in various surgical defects. PRP derives from the centrifugation of peripheral blood and has a high concentration of growth factors that promote healing. Recently, the use of adipose-derived mesenchymal stem cells (ADMSCs) has been thoroughly investigated as a form of wound healing enhancement. ADMSCs are autologous stem cells deriving from fat tissue, with a capability of differentiation in specific cells, depending on the micro-environment that they are exposed to. The aim of the present comprehensive review is to record the experimental studies that have been published and investigate the synergistic use of PRP and ADMSC in animal models. The technical aspects of experimentations, as well as the major results of each study, are discussed. In addition, the limited clinical studies including humans are also reported. Future perspectives are discussed, along with the limitations of current studies on the long-term follow up needed on efficacy and safety.
Collapse
Affiliation(s)
- Dimitris Tatsis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
- Oral and Maxillofacial Surgery Department, School of Dentistry, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece;
- Correspondence: or ; Tel.: +30-693-2611-752
| | - Varvara Vasalou
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| | - Efstathios Kotidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| | - Elissavet Anestiadou
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| | - Ioannis Grivas
- Laboratory of Anatomy, Histology & Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Angeliki Cheva
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Georgios Koliakos
- Department of Biochemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Gregory Venetis
- Oral and Maxillofacial Surgery Department, School of Dentistry, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece;
| | - Manousos-George Pramateftakis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| | - Nikolaos Ouzounidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| | - Stamatis Angelopoulos
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (V.V.); (E.K.); (E.A.); (M.-G.P.); (N.O.); (S.A.)
| |
Collapse
|
10
|
Construction of transplantable artificial vascular tissue based on adipose tissue-derived mesenchymal stromal cells by a cell coating and cryopreservation technique. Sci Rep 2021; 11:17989. [PMID: 34504254 PMCID: PMC8429436 DOI: 10.1038/s41598-021-97547-2] [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] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prevascularized artificial three-dimensional (3D) tissues are effective biomaterials for regenerative medicine. We have previously established a scaffold-free 3D artificial vascular tissue from normal human dermal fibroblasts (NHDFs) and umbilical vein-derived endothelial cells (HUVECs) by layer-by-layer cell coating technique. In this study, we constructed an artificial vascular tissue constructed by human adipose tissue-derived stromal cells (hASCs) and HUVECs (ASCVT) by a modified technique with cryopreservation. ASCVT showed a higher thickness with more dense vascular networks than the 3D tissue based on NHDFs. Correspondingly, 3D-cultured ASCs showed higher expression of several angiogenesis-related factors, including vascular endothelial growth factor-A and hepatic growth factor, compared to that of NHDFs. Moreover, perivascular cells in ASCVT were detected by pericyte markers, suggesting the differentiation of hASCs into pericyte-like cells. Subcutaneous transplantation of ASCVTs to nude mice resulted in an engraftment with anastomosis of host's vascular structures at 2 weeks after operation. In the engrafted tissue, the vascular network was surrounded by mural-like structure-forming hASCs, in which some parts developed to form vein-like structures at 4 weeks, suggesting the generation of functional vessel networks. These results demonstrated that cryopreserved human cells, including hASCs, could be used directly to construct the artificial transplantable tissue for regenerative medicine.
Collapse
|
11
|
Al-Ghadban S, Bunnell BA. Adipose Tissue-Derived Stem Cells: Immunomodulatory Effects and Therapeutic Potential. Physiology (Bethesda) 2021; 35:125-133. [PMID: 32027561 DOI: 10.1152/physiol.00021.2019] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) can self-renew and differentiate along multiple cell lineages. ASCs are also potently anti-inflammatory due to their inherent ability to regulate the immune system by secreting anti-inflammatory cytokines and growth factors that play a crucial role in the pathology of many diseases, including multiple sclerosis, diabetes mellitus, Crohn's, SLE, and graft-versus-host disease. The immunomodulatory effects and mechanisms of action of ASCs on pathological conditions are reviewed here.
Collapse
Affiliation(s)
- Sara Al-Ghadban
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bruce A Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
12
|
Kim YH, Jung E, Im GB, Kim YJ, Kim SW, Jeong GJ, Jang YC, Park KM, Kim DI, Yu T, Bhang SH. Regulation of intracellular transition metal ion level with a pH-sensitive inorganic nanocluster to improve therapeutic angiogenesis by enriching conditioned medium retrieved from human adipose derived stem cells. NANO CONVERGENCE 2020; 7:34. [PMID: 33064240 PMCID: PMC7567771 DOI: 10.1186/s40580-020-00244-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
Cell therapy based on human adipose derived stem cells (hADSCs) is a known potential therapeutic approach to induce angiogenesis in ischemic diseases. However, the therapeutic efficacy of direct hADSC injection is limited by a low cell viability and poor cell engraftment after administration. To improve the outcomes of this kind of approach, various types of nanoparticles have been utilized to improve the therapeutic efficacy of hADSC transplantation. Despite their advantages, the adverse effects of nanoparticles, such as genetic damage and potential oncogenesis based on non-degradable property of nanoparticles prohibit the application of nanoparticles toward the clinical applications. Herein, we designed a transition metal based inorganic nanocluster able of pH-selective degradation (ps-TNC), with the aim of enhancing an hADSC based treatment of mouse hindlimb ischemia. Our ps-TNC was designed to undergo degradation at low pH conditions, thus releasing metal ions only after endocytosis, in the endosome. To eliminate the limitations of both conventional hADSC injection and non-degradable property of nanoparticles, we have collected conditioned medium (CM) from the ps-TNC treated hADSCs and administrated it to the ischemic lesions. We found that intracellular increment of transition metal ion upregulated the hypoxia-inducible factor 1α, which can induce vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) expressions. Based on the molecular mechanism, the secretion of VEGF and bFGF by ps-TNC treated hADSCs showed a significant improvement compared to that of untreated cells. Injecting the CM collected from ps-TNC treated hADSCs into the mouse hindlimb ischemia model (ps-TNC-CM group) showed significantly improved angiogenesis in the lesions, with improved limb salvage and decreased muscle degeneration compared to the group injected with CM collected from normal hADSCs (CM group). This study suggests a novel strategy, combining a known angiogenesis molecular mechanism with both an improvement on conventional stem cell therapy and the circumvention of some limitations still present in modern approaches based on nanoparticles.
Collapse
Affiliation(s)
- Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Euiyoung Jung
- Department of Chemical Engineering, College of Engineering, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Gun-Jae Jeong
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Young Charles Jang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Biomedical Engineering, The Wallace H. Coulter, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Kyung Min Park
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Taekyung Yu
- Department of Chemical Engineering, College of Engineering, Kyung Hee University, Yongin, 17104, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 440-746, Republic of Korea.
| |
Collapse
|
13
|
Moussa MH, Hamam GG, Abd Elaziz AE, Rahoma MA, Abd El Samad AA, El-Waseef DAA, Hegazy MA. Comparative Study on Bone Marrow-Versus Adipose-Derived Stem Cells on Regeneration and Re-Innervation of Skeletal Muscle Injury in Wistar Rats. Tissue Eng Regen Med 2020; 17:887-900. [PMID: 33030680 DOI: 10.1007/s13770-020-00288-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Skeletal muscle injuries are frequent clinical challenges due to associated fibrosis and disability. Regenerative medicine is an emerging promising strategy for such cases. The aim of this study was to compare between the effects of bone marrow-mesenchymal stem cells (BM-MSCs) versus adipose tissue stromal cells (ADSCs) on regeneration and re-innervation of skeletal muscle laceration injury in Wistar rats at different time intervals. METHODS Six young male rats were used as a source of allogenic MSCs. Eighty-four adult female rats were divided into: Group I (control), Group II (Untreated Laceration): right gluteal muscle was lacerated and left for spontaneous healing, Group III (BM-MSCs): right gluteal muscle was lacerated with concomitant local intramuscular injection of 1 × 106 BM-MSCs in the lacerated muscle, Group IV (ADSCs): right gluteal muscle was lacerated with concomitant local intramuscular injection of 1 × 106 ADSCs in lacerated muscle. Rats were sacrificed after one, two and eight weeks. Muscles were processed to prepare sections stained with H&E, Mallory's trichrome and immune-histochemical staining (neurofilament light chain). RESULTS A significant increase in collagen fibers and failure of re-innervation were noticed in untreated laceration group. BM-MSCs-treated groups showed regeneration of muscle fibers but with increased collagen fibers. Meanwhile, ADSCs showed better regenerative effects evidenced by significant increase in the number of myotubes and significant decrease in collagen deposition. Re-innervation was noticed in MSCs-injected muscles after 8 weeks of laceration. CONCLUSION Both BM-MSCs and ADSCs improved regeneration of skeletal muscle laceration injury at short- and long-term durations. However, fibrosis was less in ADSCs-treated rats. Effective re-innervation of injured muscles occurred only at the long-term duration.
Collapse
Affiliation(s)
- Manal H Moussa
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| | - Ghada G Hamam
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt.
| | - Asmaa E Abd Elaziz
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| | - Marwa A Rahoma
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| | - Abeer A Abd El Samad
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| | - Dalia A A El-Waseef
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| | - Mohamed A Hegazy
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, El-Khalyfa El-Mamoun Street Abbasya, Cairo, Egypt
| |
Collapse
|
14
|
Lu H, Merfeld-Clauss S, Jawed Y, March KL, Coleman ME, Bogatcheva NV. Distinct Factors Secreted by Adipose Stromal Cells Protect the Endothelium From Barrier Dysfunction and Apoptosis. Front Cell Dev Biol 2020; 8:584653. [PMID: 33102487 PMCID: PMC7554254 DOI: 10.3389/fcell.2020.584653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
We have shown previously that adipose stromal cell (ASC)-derived conditioned media (CM) limited lung injury, endothelial barrier dysfunction, and apoptosis. Here, we used endothelial hyperpermeability and apoptosis assays to investigate how concentration processes affect endothelium-directed bioactivity of ASC-CM and to gain information on the nature of bioactive factors. Comparison of ASC-CM concentrated with differential molecular weight (MW) cutoff filters showed that endothelial barrier protection depended on the species-specific factors in ASC-CM fractionated with MW > 50 kDa. Known barrier regulators-keratin growth factor (KGF), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF)-were detected in ASC-CM fraction of > 100 kDa. Pretreatment of endothelial monolayers with concentrations of KGF, VEGF, and HGF detected in ASC-CM showed that only KGF and HGF protect the endothelium from barrier dysfunction. Depletion of KGF and HGF from ASC-CM attenuated ASC-CM's ability to protect the endothelial barrier. In contrast to barrier-protective factors, apoptosis-protective factors fractionated with MW < 3 kDa and were not species-specific. Application of donors of apoptosis-mitigating gases showed that the CO donor carbon monoxide-releasing molecule 2 (CORM2) protected the endothelium from apoptosis, while the H2S donor NaSH did not. Knockdown of CO-generating heme oxygenase 1 in ASC attenuated ASC-CM's ability to protect the endothelium from apoptosis. We have shown that tumor necrosis factor alpha (TNFα)-induced apoptosis in endothelium is c-Jun N-terminal kinase (JNK)-dependent, and JNK activation is inhibited by ASC-CM pretreatment of endothelial cells. ASC-CM from heme oxygenase 1-depleted ASC displayed attenuated ability to suppress endothelial JNK activation, suggesting that CO-mediated protection of the endothelium from apoptosis is achieved by the downregulation of the JNK pathway. Altogether, our results demonstrate that the concentration of ASC-CM with low MW cutoff filters significantly reduces its anti-apoptotic activity while preserving its barrier-protective activity.
Collapse
Affiliation(s)
- Hongyan Lu
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Stephanie Merfeld-Clauss
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Yameena Jawed
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Keith L March
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | | | - Natalia V Bogatcheva
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States.,Division of Pulmonary, Sleep and Critical Care, Department of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
15
|
Maleitzke T, Elazaly H, Festbaum C, Eder C, Karczewski D, Perka C, Duda GN, Winkler T. Mesenchymal Stromal Cell-Based Therapy-An Alternative to Arthroplasty for the Treatment of Osteoarthritis? A State of the Art Review of Clinical Trials. J Clin Med 2020; 9:jcm9072062. [PMID: 32630066 PMCID: PMC7409016 DOI: 10.3390/jcm9072062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disorder worldwide and to date no regenerative treatment has been established in clinical practice. This review evaluates the current literature on the clinical translation of mesenchymal stromal cell (MSC)-based therapy in OA management with a focus on safety, outcomes and procedural specifics. PubMed, Cochrane Library and clinicaltrials.gov were searched for clinical studies using MSCs for OA treatment. 290 articles were initially identified and 42 articles of interest, including a total of 1325 patients, remained for further examination. Most of the included studies used adipose tissue-derived MSCs or bone-marrow-derived MSCs to treat patients suffering from knee OA. MSC-based therapy for knee OA appears to be safe and presumably effective in selected parameters. Yet, a direct comparison between studies was difficult due to a pronounced variance regarding methodology, assessed outcomes and evidence levels. Intensive scientific engagement is needed to identify the most effective source and dosage of MSCs for OA treatment in the future. Consent on outcome measures has to be reached and eventually patient sub-populations need to be identified that will profit most from MSC-based treatment for OA.
Collapse
Affiliation(s)
- Tazio Maleitzke
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Hisham Elazaly
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Christian Festbaum
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Christian Eder
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Daniel Karczewski
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tobias Winkler
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-559084
| |
Collapse
|
16
|
Ntege EH, Sunami H, Shimizu Y. Advances in regenerative therapy: A review of the literature and future directions. Regen Ther 2020; 14:136-153. [PMID: 32110683 PMCID: PMC7033303 DOI: 10.1016/j.reth.2020.01.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/14/2020] [Accepted: 01/26/2020] [Indexed: 12/14/2022] Open
Abstract
There is enormous global anticipation for stem cell-based therapies that are safe and effective. Numerous pre-clinical studies present encouraging results on the therapeutic potential of different cell types including tissue derived stem cells. Emerging evidences in different fields of research suggest several cell types are safe, whereas their therapeutic application and effectiveness remain challenged. Multiple factors that influence treatment outcomes are proposed including immunocompatibility and potency, owing to variations in tissue origin, ex-vivo methodologies for preparation and handling of the cells. This communication gives an overview of literature data on the different types of cells that are potentially promising for regenerative therapy. As a case in point, the recent trends in research and development of the mesenchymal stem cells (MSCs) for cell therapy are considered in detail. MSCs can be isolated from a variety of tissues and organs in the human body including bone marrow, adipose, synovium, and perinatal tissues. However, MSC products from the different tissue sources exhibit unique or varied levels of regenerative abilities. The review finally focuses on adipose tissue-derived MSCs (ASCs), with the unique properties such as easier accessibility and abundance, excellent proliferation and differentiation capacities, low immunogenicity, immunomodulatory and many other trophic properties. The suitability and application of the ASCs, and strategies to improve the innate regenerative capacities of stem cells in general are highlighted among others.
Collapse
Affiliation(s)
- Edward H. Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Hiroshi Sunami
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
| |
Collapse
|
17
|
Ceccarelli S, Pontecorvi P, Anastasiadou E, Napoli C, Marchese C. Immunomodulatory Effect of Adipose-Derived Stem Cells: The Cutting Edge of Clinical Application. Front Cell Dev Biol 2020; 8:236. [PMID: 32363193 PMCID: PMC7180192 DOI: 10.3389/fcell.2020.00236] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose-derived stem cells (ASCs) represent a promising tool for soft tissue engineering as well as for clinical treatment of inflammatory and autoimmune pathologies. The well-characterized multi-differentiation potential and self-renewal properties of ASCs are coupled with their immunomodulatory ability in providing therapeutic efficacy. Yet, their impact in immune or inflammatory disorders might rely both on cell contact-dependent mechanisms and paracrine effects, resulting in the release of various soluble factors that regulate immune cells functions. Despite the widespread use of ASCs in clinical trials addressing several pathologies, the pathophysiological mechanisms at the basis of their clinical use have been not yet fully investigated. In particular, a thorough analysis of ASC immunomodulatory potential is mandatory. Here we explore such molecular mechanisms involved in ASC immunomodulatory properties, emphasizing the relevance of the milieu composition. We review the potential clinical use of ASC secretome as a mediator for immunomodulation, with a focus on in vitro and in vivo environmental conditions affecting clinical outcome. We describe some potential strategies for optimization of ASCs immunomodulatory capacity in clinical settings, which act either on adult stem cells gene expression and local microenvironment. Finally, we discuss the limitations of both allogeneic and autologous ASC use, highlighting the issues to be fixed in order to significantly improve the efficacy of ASC-based cell therapy.
Collapse
Affiliation(s)
- Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Eleni Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, Università della Campania “Luigi Vanvitelli”, Naples, Italy
- IRCCS SDN, Naples, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
18
|
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: 4] [Impact Index Per Article: 0.7] [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
|
19
|
Rowe G, Kelm NQ, Beare JE, Tracy E, Yuan F, LeBlanc AJ. Enhanced beta-1 adrenergic receptor responsiveness in coronary arterioles following intravenous stromal vascular fraction therapy in aged rats. Aging (Albany NY) 2019; 11:4561-4578. [PMID: 31296794 PMCID: PMC6660031 DOI: 10.18632/aging.102069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/25/2019] [Indexed: 04/30/2023]
Abstract
Our past study showed that a single tail vein injection of adipose-derived stromal vascular fraction (SVF) into old rats was associated with improved dobutamine-mediated coronary flow reserve. We hypothesize that i.v. injection of SVF improves coronary microvascular function in aged rats via alterations in beta adrenergic microvascular signaling. Female Fischer-344 rats aged young (3 months, n=32) and old (24 months, n=30) were utilized, along with two cell therapies intravenously injected in old rats four weeks prior to sacrifice: 1x107 green fluorescent protein (GFP+) SVF cells (O+SVF, n=21), and 5x106 GFP+ bone-marrow mesenchymal stromal cells (O+BM, n=6), both harvested from young donors. Cardiac ultrasound and pressure-volume measurements were obtained, and coronary arterioles were isolated from each group for microvessel reactivity studies and immunofluorescence staining. Coronary flow reserve decreased with advancing age, but this effect was rescued by the SVF treatment in the O+SVF group. Echocardiography showed an age-related diastolic dysfunction that was improved with SVF to a greater extent than with BM treatment. Coronary arterioles isolated from SVF-treated rats showed amelioration of the age-related decrease in vasodilation to a non-selective β-AR agonist. I.v. injected SVF cells improved β-adrenergic receptor-dependent coronary flow and microvascular function in a model of advanced age.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| | - Natia Q. Kelm
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
| | - Jason E. Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA
| | - Evan Tracy
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| | - Fangping Yuan
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
| | - Amanda J. LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40292, USA
- Department of Physiology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
20
|
Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci 2019; 20:ijms20123105. [PMID: 31242644 PMCID: PMC6627452 DOI: 10.3390/ijms20123105] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic musculoskeletal (MSK) pain is one of the most common medical complaints worldwide and musculoskeletal injuries have an enormous social and economical impact. Current pharmacological and surgical treatments aim to relief pain and restore function; however, unsatiscactory outcomes are commonly reported. In order to find an accurate treatment to such pathologies, over the last years, there has been a significantly increasing interest in cellular therapies, such as adipose-derived mesenchymal stem cells (AMSCs). These cells represent a relatively new strategy in regenerative medicine, with many potential applications, especially regarding MSK disorders, and preclinical and clinical studies have demonstrated their efficacy in muscle, tendon, bone and cartilage regeneration. Nevertheless, several worries about their safety and side effects at long-term remain unsolved. This article aims to review the current state of AMSCs therapy in the treatment of several MSK diseases and their clinical applications in veterinary and human medicine.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
21
|
Plotkin JD, Elias MG, Fereydouni M, Daniels-Wells TR, Dellinger AL, Penichet ML, Kepley CL. Human Mast Cells From Adipose Tissue Target and Induce Apoptosis of Breast Cancer Cells. Front Immunol 2019; 10:138. [PMID: 30833944 PMCID: PMC6387946 DOI: 10.3389/fimmu.2019.00138] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/16/2019] [Indexed: 01/09/2023] Open
Abstract
Mast cells (MC) are important immune sentinels found in most tissue and widely recognized for their role as mediators of Type I hypersensitivity. However, they also secrete anti-cancer mediators such as tumor necrosis factor alpha (TNF-α) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The purpose of this study was to investigate adipose tissue as a new source of MC in quantities that could be used to study MC biology focusing on their ability to bind to and kill breast cancer cells. We tested several cell culture media previously demonstrated to induce MC differentiation. We report here the generation of functional human MC from adipose tissue. The adipose-derived mast cells (ADMC) are phenotypically and functionally similar to connective tissue expressing tryptase, chymase, c-kit, and FcεRI and capable of degranulating after cross-linking of FcεRI. The ADMC, sensitized with anti-HER2/neu IgE antibodies with human constant regions (trastuzumab IgE and/or C6MH3-B1 IgE), bound to and released MC mediators when incubated with HER2/neu-positive human breast cancer cells (SK-BR-3 and BT-474). Importantly, the HER2/neu IgE-sensitized ADMC induced breast cancer cell (SK-BR-3) death through apoptosis. Breast cancer cell apoptosis was observed after the addition of cell-free supernatants containing mediators released from FcεRI-challenged ADMC. Apoptosis was significantly reduced when TNF-α blocking antibodies were added to the media. Adipose tissue represents a source MC that could be used for multiple research purposes and potentially as a cell-mediated cancer immunotherapy through the expansion of autologous (or allogeneic) MC that can be targeted to tumors through IgE antibodies recognizing tumor specific antigens.
Collapse
Affiliation(s)
- Jesse D Plotkin
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Michael G Elias
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Mohammad Fereydouni
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anthony L Dellinger
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.,AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States.,The California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher L Kepley
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| |
Collapse
|
22
|
Argentati C, Morena F, Bazzucchi M, Armentano I, Emiliani C, Martino S. Adipose Stem Cell Translational Applications: From Bench-to-Bedside. Int J Mol Sci 2018; 19:E3475. [PMID: 30400641 PMCID: PMC6275042 DOI: 10.3390/ijms19113475] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023] Open
Abstract
During the last five years, there has been a significantly increasing interest in adult adipose stem cells (ASCs) as a suitable tool for translational medicine applications. The abundant and renewable source of ASCs and the relatively simple procedure for cell isolation are only some of the reasons for this success. Here, we document the advances in the biology and in the innovative biotechnological applications of ASCs. We discuss how the multipotential property boosts ASCs toward mesenchymal and non-mesenchymal differentiation cell lineages and how their character is maintained even if they are combined with gene delivery systems and/or biomaterials, both in vitro and in vivo.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Ilaria Armentano
- Department of Ecological and Biological Sciences, Tuscia University Largo dell'Università, snc, 01100 Viterbo, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
23
|
Lis-Bartos A, Smieszek A, Frańczyk K, Marycz K. Fabrication, Characterization, and Cytotoxicity of Thermoplastic Polyurethane/Poly(lactic acid) Material Using Human Adipose Derived Mesenchymal Stromal Stem Cells (hASCs). Polymers (Basel) 2018; 10:E1073. [PMID: 30960998 PMCID: PMC6403585 DOI: 10.3390/polym10101073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/15/2022] Open
Abstract
Thermoplastic polyurethane (TPU) and poly(lactic acid) are types of biocompatible and degradable synthetic polymers required for biomedical applications. Physically blended (TPU+PLA) tissue engineering matrices were produced via solvent casting technique. The following types of polymer blend were prepared: (TPU+PLA) 7:3, (TPU+PLA) 6:4, (TPU+PLA) 4:6, and (TPU+PLA) 3:7. Various methods were employed to characterize the properties of these polymers: surface properties such as morphology (scanning electron microscopy), wettability (goniometry), and roughness (profilometric analysis). Analyses of hydrophilic and hydrophobic properties, thermogravimetric analysis (TGA), and differential scanning calorimetry (DSC) of the obtained polymer blends were conducted. Tensile tests demonstrated that the blends exhibited a wide range of mechanical properties. Cytotoxicity of polymers was tested using human multipotent stromal cells derived from adipose tissue (hASC). In vitro assays revealed that (TPU+PLA) 3:7 matrices were the most cytocompatible biomaterials. Cells cultured on (TPU+PLA) 3:7 had proper morphology, growth pattern, and were distinguished by increased proliferative and metabolic activity. Additionally, it appeared that (TPU+PLA) 3:7 biomaterials showed antiapoptotic properties. hASC cultured on these matrices had reduced expression of Bax-α and increased expression of Bcl-2. This study demonstrated the feasibility of producing a biocompatible scaffold form based on (TPU+PLA) blends that have potential to be applied in tissue engineering.
Collapse
Affiliation(s)
- Anna Lis-Bartos
- AGH University of Science and Technology, Department of Biomaterials and Composites, Faculty of Materials Science and Science and Ceramics, Krakow 30-059, Poland.
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw 50-375, Poland.
| | - Agnieszka Smieszek
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw 50-375, Poland.
| | - Kinga Frańczyk
- AGH University of Science and Technology, Faculty of Electrical Engineering, Automatics, Computer Science and Biomedical Engineering, Krakow 30-059, Poland.
| | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw 50-375, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, Gießen 35392, Germany.
| |
Collapse
|
24
|
Kelm NQ, Beare JE, Yuan F, George M, Shofner CM, Keller BB, Hoying JB, LeBlanc AJ. Adipose-derived cells improve left ventricular diastolic function and increase microvascular perfusion in advanced age. PLoS One 2018; 13:e0202934. [PMID: 30142193 PMCID: PMC6108481 DOI: 10.1371/journal.pone.0202934] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
An early manifestation of coronary artery disease in advanced age is the development of microvascular dysfunction leading to deficits in diastolic function. Our lab has previously shown that epicardial treatment with adipose-derived stromal vascular fraction (SVF) preserves microvascular function following coronary ischemia in a young rodent model. Follow-up studies showed intravenous (i.v.) delivery of SVF allows the cells to migrate to the walls of small vessels and reset vasomotor tone. Therefore we tested the hypothesis that the i.v. cell injection of SVF would reverse the coronary microvascular dysfunction associated with aging in a rodent model. Fischer 344 rats were divided into 4 groups: young control (YC), old control (OC), old + rat aortic endothelial cells (O+EC) and old + GFP+ SVF cells (O+SVF). After four weeks, cardiac function and coronary flow reserve (CFR) were measured via echocardiography, and hearts were explanted either for histology or isolation of coronary arterioles for vessel reactivity studies. In a subgroup of animals, microspheres were injected during resting and dobutamine-stimulated conditions to measure coronary blood flow. GFP+ SVF cells engrafted and persisted in the myocardium and coronary vasculature four weeks following i.v. injection. Echocardiography showed age-related diastolic dysfunction without accompanying systolic dysfunction; diastolic function was improved in old rats after SVF treatment. Ultrasound and microsphere data both showed increased stimulated coronary blood flow in O+SVF rats compared to OC and O+EC, while isolated vessel reactivity was mostly unchanged. I.v.-injected SVF cells were capable of incorporating into the vasculature of the aging heart and are shown in this study to improve CFR and diastolic function in a model of advanced age. Importantly, SVF injection did not lead to arrhythmias or increased mortality in aged rats. SVF cells provide an autologous cell therapy option for treatment of microvascular and cardiac dysfunction in aged populations.
Collapse
Affiliation(s)
- Natia Q. Kelm
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Jason E. Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States of America
| | - Fangping Yuan
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Monika George
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Charles M. Shofner
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Bradley B. Keller
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Pediatrics, University of Louisville, Louisville, Kentucky, United States of America
| | - James B. Hoying
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Amanda J. LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
25
|
Yin Y, Chen P, Yu Q, Peng Y, Zhu Z, Tian J. The Effects of a Pulsed Electromagnetic Field on the Proliferation and Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Med Sci Monit 2018; 24:3274-3282. [PMID: 29775452 PMCID: PMC5987610 DOI: 10.12659/msm.907815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background A low frequency pulsed electromagnetic field (PEMF) has been confirmed to play an important role in promoting the osteogenic differentiation of human bone marrow stem cells (BMSCs). Adipose-derived stem cells (ASCs) possess some attractive characteristics for clinical application compared to BMSCs, such as abundant stem cells from lipoaspirates, faster growth, less discomfort and morbidity during surgery. ASCs can become adipocytes, osteoblasts, chondrocytes, myocytes, neurocytes, and other cell types. Thus, ASCs might be a good alternative in clinical work involving treatment with PEMF. Material/Methods Human ASCs (hASCs)were divided into a control group (without PEMF exposure) and an experimental group (PEMF for two hours per day). We examined the effect of PEMF on promoting cell proliferation and osteogenic differentiation from several aspects: CCK-8 proliferation assay, RNA extraction, qRT-PCR detection, western blotting, and immunofluorescence staining experiments. Results PEMF could promote cell proliferation of human ASCs (hASCs) at an early stage as determined by CCK-8 assay. A specific intensity (1 mT) and frequency (50 Hz) of PEMF promoted osteogenic differentiation in hASCs in alkaline phosphatase (ALP) staining experiments. In addition, bone-related gene expression increased after two weeks of PEMF exposure, the protein expression of OPN, OCN, and RUNX-2 also increased after a longer period (three weeks) of PEMF treatment as determined by western blotting and immunofluorescence staining. Conclusions We found for the first time that PMEF has a role in stimulating cell proliferation of hASCs at an early period, subsequently promoting bone-related gene expression and inducing the expression of related proteins to stimulate osteogenic differentiation.
Collapse
Affiliation(s)
- Yukun Yin
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Ping Chen
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Qiang Yu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Yan Peng
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Baiyun, Guangzhou, China (mainland)
| | - ZeHao Zhu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Jing Tian
- Department of Orthopedics, Zhujiang Hospital,Southern Medical University, Haizhu, Guangzhou, China (mainland)
| |
Collapse
|
26
|
Miana VV, González EAP. Adipose tissue stem cells in regenerative medicine. Ecancermedicalscience 2018; 12:822. [PMID: 29662535 PMCID: PMC5880231 DOI: 10.3332/ecancer.2018.822] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Indexed: 12/26/2022] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) are mesenchymal cells with the capacity for self-renewal and multipotential differentiation. This multipotentiality allows them to become adipocytes, chondrocytes, myocytes, osteoblasts and neurocytes among other cell lineages. Stem cells and, in particular, adipose tissue-derived cells, play a key role in reconstructive or tissue engineering medicine as they have already proven effective in developing new treatments. The purpose of this work is to review the applications of ADSCs in various areas of regenerative medicine, as well as some of the risks associated with treatment with ADSCs in neoplastic disease.
Collapse
Affiliation(s)
- Vanesa Verónica Miana
- Centre for Advanced Studies in Humanities and Health Sciences, Interamerican Open University, Buenos Aires, Argentina
| | - Elio A Prieto González
- Centre for Advanced Studies in Humanities and Health Sciences, Interamerican Open University, Buenos Aires, Argentina
| |
Collapse
|
27
|
Current Therapeutic Strategies for Stem Cell-Based Cartilage Regeneration. Stem Cells Int 2018; 2018:8490489. [PMID: 29765426 PMCID: PMC5889878 DOI: 10.1155/2018/8490489] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/14/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
The process of cartilage destruction in the diarthrodial joint is progressive and irreversible. This destruction is extremely difficult to manage and frustrates researchers, clinicians, and patients. Patients often take medication to control their pain. Surgery is usually performed when pain becomes uncontrollable or joint function completely fails. There is an unmet clinical need for a regenerative strategy to treat cartilage defect without surgery due to the lack of a suitable regenerative strategy. Clinicians and scientists have tried to address this using stem cells, which have a regenerative potential in various tissues. Cartilage may be an ideal target for stem cell treatment because it has a notoriously poor regenerative potential. In this review, we describe past, present, and future strategies to regenerate cartilage in patients. Specifically, this review compares a surgical regenerative technique (microfracture) and cell therapy, cell therapy with and without a scaffold, and therapy with nonaggregated and aggregated cells. We also review the chondrogenic potential of cells according to their origin, including autologous chondrocytes, mesenchymal stem cells, and induced pluripotent stem cells.
Collapse
|
28
|
D'Ambrosi R, Indino C, Maccario C, Manzi L, Usuelli FG. Autologous Microfractured and Purified Adipose Tissue for Arthroscopic Management of Osteochondral Lesions of the Talus. J Vis Exp 2018. [PMID: 29443020 DOI: 10.3791/56395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In recent years, regenerative techniques have been increasingly studied and used to treat osteochondral lesions of the talus. In particular, several studies have focused their attention on mesenchymal stem cells derived from adipose tissue. Adipose-derived stem cells (ADSCs) exhibit morphological characteristics and properties similar to other mesenchymal cells, and are able to differentiate into several cellular lines. Moreover, these cells are also widely available in the subcutaneous tissue, representing 10 - 30% of the normal body weight, with a concentration of 5,000 cells per gram of tissue. In the presented technique, the first step involves harvesting ADSCs from the abdomen and a process of microfracture and purification; next, the surgical procedure is performed entirely arthroscopically, with less soft tissue dissection, better joint visualization, and a faster recovery compared with standard open procedures. Arthroscopy is characterized by a first phase in which the lesion is identified, isolated, and prepared with microperforations; the second step, performed dry, involves injection of adipose tissue at the level of the lesion. Between January 2016 and September 2016, four patients underwent arthroscopic treatment of osteochondral lesion of the talus with microfractured and purified adipose tissue. All patients reported clinical improvement six months after surgery with no reported complications. Functional scores at the latest follow-up are encouraging and confirm that the technique provides reliable pain relief and improvements in patients with osteochondral lesion of the talus.
Collapse
Affiliation(s)
- Riccardo D'Ambrosi
- IRCCS Istituto Ortopedico Galeazzi - C.A.S.C.O. Piede e Caviglia; Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano;
| | - Cristian Indino
- IRCCS Istituto Ortopedico Galeazzi - C.A.S.C.O. Piede e Caviglia
| | - Camilla Maccario
- IRCCS Istituto Ortopedico Galeazzi - C.A.S.C.O. Piede e Caviglia
| | - Luigi Manzi
- IRCCS Istituto Ortopedico Galeazzi - C.A.S.C.O. Piede e Caviglia
| | | |
Collapse
|
29
|
Xie H, Liao N, Lan F, Cai Z, Liu X, Liu J. 3D-cultured adipose tissue-derived stem cells inhibit liver cancer cell migration and invasion through suppressing epithelial-mesenchymal transition. Int J Mol Med 2017; 41:1385-1396. [PMID: 29286072 PMCID: PMC5819936 DOI: 10.3892/ijmm.2017.3336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/13/2017] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) are considered promising candidates for stem cell therapy; however, the tumorigenicity of ADSCs remains controversial. The present study aimed to investigate the association between ADSCs and liver cancer cells, and to determine whether culture methods could influence the effects of ADSCs on liver cancer cell growth in vitro. Liver cancer cells were treated with ADSCs-conditioned medium (CM) that was collected using the two-dimensional (2D) culture method, sphere culture method, or three-dimensional (3D) culture method. After that, cell viability and apoptosis were measured using CCK-8 and Annexin V-FITC assay, respectively; the cell motility and adhesive capacity were analyzed by scratch wound healing and cell adhesion assay, respectively; the cell migration and invasion were examined by Transwell units; and the molecular mechanisms of ADSCs on effecting epithelial mesenchymal transition signaling pathway were further analyzed. The results demonstrated that ADSCs-CM was able to inhibit the growth of liver cancer cells by inhibiting cell proliferation and promoting cell apoptosis, as well as by suppressing cell motility, adhesive capacity, migration and invasion. In addition, ADSCs-CM was able to suppress cell growth via the downregulation of epithelial-mesenchymal transition signaling. Notably, the enhanced inhibitory effects of ADSCs on liver cancer cell growth could be achieved after cultu ring using a 3D approach. These findings suggested that ADSCs may provide a novel promising therapeutic approach for the treatment of patients with liver cancer, and the 3D culture method may provide a novel approach to explore the association between ADSCs and cancer.
Collapse
Affiliation(s)
- Haihua Xie
- Department of Clinical Genetics and Experimental Medicine, Fuzong Clinical College, Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Fenghua Lan
- Department of Clinical Genetics and Experimental Medicine, Fuzong Clinical College, Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
30
|
De novo lipogenesis and desaturation of fatty acids during adipogenesis in bovine adipose-derived mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2017; 54:23-31. [PMID: 29192407 DOI: 10.1007/s11626-017-0205-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/06/2017] [Indexed: 10/18/2022]
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are useful cell model to study adipogenesis and energy metabolism. However, the biological characteristics of bovine ADSCs (bADSCs) remain unclear. This study aimed to isolate and identify bADSCs and further investigate fatty acid (FA)-related gene expression and composition of FAs during adipogenesis. The growth curve showed the bADSCs of P5 cells had rapid proliferation superior to P10-P50. The colony formation assay showed colony number of P5 cells was higher than that of P50 cells (51.67 ± 3.06 vs 35.67 ± 6.43, P < 0.05). The immunofluorescence showed that bADSCs were positive for CD13, CD44, CD49d, CD90, CD105, and Vimentin while negative for CD34. The multipotential towards adipocyte, osteocyte, and chondrocyte was confirmed by specific histological staining and lineage gene expression. During adipogenic induction, the genes related to lipogenesis and lipolysis were assessed by real-time PCR and the FA composition was detected by GC-MS. Expression of lipogenesis-related genes showed coordinated regulation as peaking on day 7 and declining until induction ended, including PPARγ, SREBP1, ACC1, FAS, ELOVL6, SCD1, and FABP4. FA deposition-related genes (DGAT1 and ACAT1) increased until day 14. Lipolysis genes (CPT-1A, VLCAD, and ACO) showed a variant expression pattern. The profile of FAs showed that proportion of the FAs (C4-C15, ≥ C22) increased, but proportion of long-chain fatty acids (C16-C20) reduced after induction. And saturated FAs (SFA) decreased while monounsaturated FAs (MUFA) and polyunsaturated FAs (PUFA) increased during adipogenesis. These data suggest that bADSCs possess the characteristics of mesenchymal stem cells and have active de novo lipogenesis (DNL) and desaturation of FAs during adipogenesis.
Collapse
|
31
|
Tabatabaei Qomi R, Sheykhhasan M. Adipose-derived stromal cell in regenerative medicine: A review. World J Stem Cells 2017; 9:107-117. [PMID: 28928907 PMCID: PMC5583529 DOI: 10.4252/wjsc.v9.i8.107] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 05/19/2017] [Indexed: 02/06/2023] Open
Abstract
The application of appropriate cell origin for utilizing in regenerative medicine is the major issue. Various kinds of stem cells have been used for the tissue engineering and regenerative medicine. Such as, several stromal cells have been employed as treat option for regenerative medicine. For example, human bone marrow-derived stromal cells and adipose-derived stromal cells (ADSCs) are used in cell-based therapy. Data relating to the stem cell therapy and processes associated with ADSC has developed remarkably in the past 10 years. As medical options, both the stromal vascular and ADSC suggests good opportunity as marvelous cell-based therapeutics. The some biological features are the main factors that impact the regenerative activity of ADSCs, including the modulation of the cellular immune system properties and secretion of bioactive proteins such as cytokines, chemokines and growth factors, as well as their intrinsic anti-ulcer and anti-inflammatory potential. A variety of diseases have been treated by ADSCs, and it is not surprising that there has been great interest in the possibility that ADSCs might be used as therapeutic strategy to improve a wider range of diseases. This is especially important when it is remembered that routine therapeutic methods are not completely effective in treat of diseases. Here, it was discuss about applications of ADSC to colitis, liver failure, diabetes mellitus, multiple sclerosis, orthopaedic disorders, hair loss, fertility problems, and salivary gland damage.
Collapse
Affiliation(s)
- Reza Tabatabaei Qomi
- Department of Stem Cell, the Academic Center for Education, Culture and Research, PO Box QOM-3713189934, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Stem Cell, the Academic Center for Education, Culture and Research, PO Box QOM-3713189934, Qom, Iran
| |
Collapse
|
32
|
Red (660 nm) or near-infrared (810 nm) photobiomodulation stimulates, while blue (415 nm), green (540 nm) light inhibits proliferation in human adipose-derived stem cells. Sci Rep 2017; 7:7781. [PMID: 28798481 PMCID: PMC5552860 DOI: 10.1038/s41598-017-07525-w] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/29/2017] [Indexed: 12/26/2022] Open
Abstract
We previously showed that blue (415 nm) and green (540 nm) wavelengths were more effective in stimulating osteoblast differentiation of human adipose-derived stem cells (hASC), compared to red (660 nm) and near-infrared (NIR, 810 nm). Intracellular calcium was higher after blue/green, and could be inhibited by the ion channel blocker, capsazepine. In the present study we asked what was the effect of these four wavelengths on proliferation of the hASC? When cultured in proliferation medium there was a clear difference between blue/green which inhibited proliferation and red/NIR which stimulated proliferation, all at 3 J/cm2. Blue/green reduced cellular ATP, while red/NIR increased ATP in a biphasic manner. Blue/green produced a bigger increase in intracellular calcium and reactive oxygen species (ROS). Blue/green reduced mitochondrial membrane potential (MMP) and lowered intracellular pH, while red/NIR had the opposite effect. Transient receptor potential vanilloid 1 (TRPV1) ion channel was expressed in hADSC, and the TRPV1 ligand capsaicin (5uM) stimulated proliferation, which could be abrogated by capsazepine. The inhibition of proliferation caused by blue/green could also be abrogated by capsazepine, and by the antioxidant, N-acetylcysteine. The data suggest that blue/green light inhibits proliferation by activating TRPV1, and increasing calcium and ROS.
Collapse
|
33
|
Arrizabalaga JH, Nollert MU. Properties of porcine adipose-derived stem cells and their applications in preclinical models. Adipocyte 2017; 6:217-223. [PMID: 28410000 DOI: 10.1080/21623945.2017.1312040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adipose-derived stem cells represent a reliable adult stem cell source thanks to their abundance, straightforward isolation, and broad differentiation abilities. Consequently, human adipose-derived stem cells (hASCs) have been used in vitro for several innovative cellular therapy and regenerative medicine applications. However, the translation of a novel technology from the laboratory to the clinic requires first to evaluate its safety, feasibility, and potential efficacy through preclinical studies in animals. The anatomy and physiology of pigs and humans are very similar, establishing pigs as an attractive and popular large animal model for preclinical studies. Knowledge of the properties of porcine adipose-derived stem cells (pASCs) used in preclinical studies is critical for their success. While hASCs have been extensively studied this past decade, only a handful of reports relate to pASCs. The aim of this concise review is to summarize the current findings about the isolation of pASCs, their culture, proliferation, and immunophenotype. The differentiation abilities of pASCs and their applications in porcine preclinical models will also be reported.
Collapse
Affiliation(s)
| | - Matthias U. Nollert
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- School of Chemical, Biological & Materials Engineering, University of Oklahoma, Norman, OK, USA
| |
Collapse
|
34
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
35
|
Bertone AL, Reisbig NA, Kilborne AH, Kaido M, Salmanzadeh N, Lovasz R, Sizemore JL, Scheuermann L, Kopp RJ, Zekas LJ, Brokken MT. Equine Dental Pulp Connective Tissue Particles Reduced Lameness in Horses in a Controlled Clinical Trial. Front Vet Sci 2017; 4:31. [PMID: 28344975 PMCID: PMC5344919 DOI: 10.3389/fvets.2017.00031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/20/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To assess if injection of allogeneic dental pulp tissue particles would improve lameness in horses with naturally occurring osteoarthritis (OA) or soft tissue (ST) injury. DESIGN Prospective, randomized, blinded, and controlled clinical trial and client survey assessment. ANIMALS Forty lame client-owned horses. PROCEDURES Sterile dental pulp, recovered from otherwise healthy foals that perish during dystocia, was processed under good manufacturing processing to produce mechanically manipulated, unexpanded pulp tissue particles containing viable cells surrounded in extracellular matrix. Forty lame client-owned horses with confirmed OA (n = 20), or ST injury (desmitis or tendonitis) received a 2 mL intra-articular (n = 20 OA) or intra-lesional (n = 20) injection of control transport vehicle (n = 20) or 10 × 106 dental pulp tissue particles (n = 20). Acclimatized horses had baseline measurements performed and were then injected on day 0. Horses were treadmill exercised for 2 weeks, evaluated by clinical parameters, lameness score, edema (score and circumference), pain on flexion (OA) or pressure (ST), and clients' scores for pain and discomfort before and through 45 days after pulp injection. Twenty horses were available for >2.5-year follow-up. RESULTS Pulp-treated horses showed decrease in lameness compared to baseline (P < 0.009) or placebo controls (P < 0.013) for at least 2 weeks. Client assessments of comfort were improved between before and 45 days after pulp injection (P < 0.001). Clinical improvement with ST injury was significantly greater than OA (P < 0.001). At >2.5-year follow-up, at least 10 horses were in work. CONCLUSION AND CLINICAL RELEVANCE Dental pulp tissue particles can be considered as a treatment option for equine lameness due to OA, desmitis, or tendonitis.
Collapse
Affiliation(s)
- Alicia L. Bertone
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Nathalie A. Reisbig
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Allison H. Kilborne
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Mari Kaido
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Navid Salmanzadeh
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Rebecca Lovasz
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Joy L. Sizemore
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Logan Scheuermann
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Rosalind J. Kopp
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Lisa J. Zekas
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Matthew T. Brokken
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
36
|
Chen YS, Hsueh YS, Chen YY, Lo CY, Tai HC, Lin FH. Evaluation of a laminin-alginate biomaterial, adipocytes, and adipocyte-derived stem cells interaction in animal autologous fat grafting model using 7-Tesla magnetic resonance imaging. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:18. [PMID: 28000114 DOI: 10.1007/s10856-016-5826-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/29/2016] [Indexed: 06/06/2023]
Abstract
Biomaterials are often added to autologous fat grafts both as supporting matrices for the grafted adipocytes and as cell carrier for adipose-derived stem cells (ADSCs). This in vivo study used an autologous fat graft model to test a lamininalginate biomaterial, adipocytes, and ADSCs in immune-competent rats. We transplanted different combinations of shredded autologous adipose tissue [designated "A" for adipose tissue]), laminin-alginate beads [designated "B" for bead], and ADSCs [designated "C" for cell]) into the backs of 15 Sprague-Dawley rats. Group A received only adipocytes, Group B received only laminin-alginate beads, Group AB received adipocytes mixed with laminin-alginate beads, Group BC received laminin-alginate beads encapsulating ADSCs, and Group ABC received adipocytes and laminin-alginate beads containing ADSCs. Seven-tesla magnetic resonance imaging was used to evaluate the rats at the 1st, 6th, and 12th weeks after transplantation. At the 12th week, the rats were sacrificed and the implanted materials were retrieved for gross examination and histological evaluation. The results based on MRI, gross evaluation, and histological data all showed that implants in Group ABC had better resorption of the biomaterial, improved survival of the grafted adipocytes, and adipogenic differentiation of ADSCs. Volume retention of grafts in Group ABC (89%) was also significantly greater than those in Group A (58%) (p < 0.01). Our findings support that the combination of shredded adipose tissue with ADSCs in laminin-alginate beads provided the best overall outcome.
Collapse
Affiliation(s)
- Yo-Shen Chen
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan
- Department of Plastic Surgery, Far Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Yu-Sheng Hsueh
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan
| | - Yen-Yu Chen
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan
| | - Cheng-Yu Lo
- Department of Pathology, Far Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Hao-Chih Tai
- Department of Plastic Surgery, National Taiwan University Hospital, Taipei, 10051, Taiwan
| | - Feng-Huei Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan.
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, 35053, Taiwan.
| |
Collapse
|
37
|
Ma T, Luan SL, Huang H, Sun XK, Yang YM, Zhang H, Han WD, Li H, Han Y. Upregulation of CC Chemokine Receptor 7 (CCR7) Enables Migration of Xenogeneic Human Adipose-Derived Mesenchymal Stem Cells to Rat Secondary Lymphoid Organs. Med Sci Monit 2016; 22:5206-5217. [PMID: 28035134 PMCID: PMC5221418 DOI: 10.12659/msm.902690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background CC chemokine receptor 7 (CCR7) expression is vital for cell migration to secondary lymphoid organs (SLOs). Our previous work showed that inducing CCR7 expression enabled syngeneic mesenchymal stem cells (MSCs) to migrate into SLOs, resulting in enhanced immunosuppressive performance in mice. Given that human adipose-derived stem cells (hASCs) are widely used in clinical therapy, we further investigated whether upregulation of CCR7 enables xenogeneic hASCs to migrate to rat SLOs. Material/Methods hASCs rarely express CCR7; therefore, hASCs were transfected with lentivirus encoding rat CCR7 (rCCR7) plus green fluorescence protein (GFP) or GFP alone. CCR7 mRNA and cell surface expression of rCCR7-hASCs and GFP-hASCs were examined by reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry (FCM), respectively. The phenotype, differentiation, and proliferation capacity of each cell type was also determined. To examine migration, rCCR7-hASCs and GFP-hASCs were injected intravenously into Lewis rats, and the proportion of GFP-positive cells in the spleen and lymph nodes was determined with FCM. Results mRNA and cell surface protein expression of CCR7 was essentially undetectable in hASCs and GFP-ASCs; however, CCR7 was highly expressed in rCCR7-ASCs. rCCR7-hASCs, GFP-hASCs, and hASCs shared a similar immunophenotype, and maintained the ability of multilineage differentiation and proliferation. In addition, the average proportion of GFP-positive cells was significantly higher following transplantation of rCCR7-hASCs compared with GFP-hASCs (p<0.01). Conclusions These results suggest that upregulation of rat CCR7 expression does not change the phenotype, differentiation, or proliferation capacity of hASCs, but does enable efficient migration of hASCs to rat SLOs.
Collapse
Affiliation(s)
- Tian Ma
- Department of Plastic and Reconstruction, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Shao-Liang Luan
- Department of Vascular and Endovascular, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hong Huang
- Institute of Basic Medicine, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Xing-Kun Sun
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, China (mainland)
| | - Yan-Mei Yang
- Department of Stomatology, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hui Zhang
- Department of Plastic Surgery, The Second Hospital of ShanXi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Wei-Dong Han
- Department of Molecular Biology, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hong Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, China (mainland)
| | - Yan Han
- Department of Plastic and Reconstruction, Chinese PLA General Hospital, Beijing, China (mainland)
| |
Collapse
|
38
|
In Vitro Characterization of Human Mesenchymal Stem Cells Isolated from Different Tissues with a Potential to Promote Complex Bone Regeneration. Stem Cells Int 2016; 2016:3595941. [PMID: 27999599 PMCID: PMC5143785 DOI: 10.1155/2016/3595941] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/19/2016] [Indexed: 01/09/2023] Open
Abstract
Bone tissue regeneration is a major, worldwide medical need, and several strategies have been developed to support the regeneration of extensive bone defects, including stem cell based bone grafts. In addition to the application of stem cells with high osteogenic potential, it is important to maintain proper blood flow in a bone graft to avoid inner graft necrosis. Mesenchymal stem cells (MSCs) may form both osteocytes and endothelial cells; therefore we examined the combined in vitro osteogenic and endothelial differentiation capacities of MSCs derived from adipose tissue, Wharton's jelly, and periodontal ligament. Based on a detailed characterization presented here, MSCs isolated from adipose tissue and periodontal ligament may be most appropriate for generating vascularized bone grafts.
Collapse
|
39
|
Nishimura A, Kumagai T, Nakatani M, Yoshimura K. Method for selective quantification of adipose-derived stromal/stem cells in tissue. J Biol Methods 2016; 3:e58. [PMID: 31453220 PMCID: PMC6706120 DOI: 10.14440/jbm.2016.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/28/2016] [Accepted: 09/26/2016] [Indexed: 12/02/2022] Open
Abstract
Fat grafts are valuable for soft-tissue regeneration and augmentation. However, fat graft systems require further improvement for the prediction of graft retention. The concentration of adipose-derived stromal/stem cells (ASCs) is one of the most important factors that affect graft retention; however, current cell quantification techniques have not been applied to adipose tissue. Here we developed a method for the selective quantification of ASCs in tissue (SQAT). We identified a characteristic methylated site in the CD31 promoter after searching for specific markers of ASCs. This DNA methylation was not detected in any cell type other than ASCs in adipose tissue. Therefore, analyzing this methylation may be a suitable approach for quantifying ASCs in tissues because DNA is readily extracted from tissues. SQAT is based on quantifying this methylation by quantitative polymerase chain reaction using methylation-sensitive HapII-treated DNA as the template. SQAT was validated based on the numbers of ASCs determined by CD31−/CD34+-based flow cytometry. The results obtained by both methods were perfectly correlated, thereby demonstrating that SQAT is a useful tool for quantifying ASCs. SQAT analysis using ASCs isolated from suctioned fat according to the standard protocol (i.e., collagenase treatment) showed that the yield of ASCs was 59% ± 21%, which suggests that the ASC isolation technique requires further improvement. Furthermore, SQAT is an excellent method for quantifying ASCs in arbitrary samples (particularly tissue), which could dramatically improve ASC isolation technologies and fat graft systems, thereby facilitating the prediction of graft retention.
Collapse
Affiliation(s)
- Akira Nishimura
- Kaneka Corporation, Kobe MI R&D Center F 6-7-3, Minatojima, Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takeo Kumagai
- Kaneka Corporation, Kobe MI R&D Center F 6-7-3, Minatojima, Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masaru Nakatani
- Kaneka Corporation, Kobe MI R&D Center F 6-7-3, Minatojima, Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kotaro Yoshimura
- Department of Plastic Surgery, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
40
|
Wang Y, Huang YY, Wang Y, Lyu P, Hamblin MR. Photobiomodulation (blue and green light) encourages osteoblastic-differentiation of human adipose-derived stem cells: role of intracellular calcium and light-gated ion channels. Sci Rep 2016; 6:33719. [PMID: 27650508 PMCID: PMC5030629 DOI: 10.1038/srep33719] [Citation(s) in RCA: 375] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022] Open
Abstract
Human adipose-derived stem cells (hASCs) have the potential to differentiate into several different cell types including osteoblasts. Photobiomodulation (PBM) or low level laser therapy (LLLT) using red or near-infrared wavelengths has been reported to have effects on both proliferation and osteogenic differentiation of stem cells. We examined the effects of delivering four different wavelengths (420 nm, 540 nm, 660 nm, 810 nm) at the same dose (3 J/cm2) five times (every two days) on hASCs cultured in osteogenic medium over three weeks. We measured expression of the following transcription factors by RT-PCR: RUNX2, osterix, and the osteoblast protein, osteocalcin. The 420 nm and 540 nm wavelengths were more effective in stimulating osteoblast differentiation compared to 660 nm and 810 nm. Intracellular calcium was higher after 420 nm and 540 nm, and could be inhibited by capsazepine and SKF96365, which also inhibited osteogenic differentiation. We hypothesize that activation of light-gated calcium ion channels by blue and green light could explain our results.
Collapse
Affiliation(s)
- Yuguang Wang
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China.,Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying-Ying Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Wang
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Peijun Lyu
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
41
|
Nitkin CR, Bonfield TL. Concise Review: Mesenchymal Stem Cell Therapy for Pediatric Disease: Perspectives on Success and Potential Improvements. Stem Cells Transl Med 2016; 6:539-565. [PMID: 28191766 PMCID: PMC5442806 DOI: 10.5966/sctm.2015-0427] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/04/2016] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a potentially revolutionary therapy for a wide variety of pediatric diseases, but the optimal cell-based therapeutics for such diversity have not yet been specified. The published clinical trials for pediatric pulmonary, cardiac, orthopedic, endocrine, neurologic, and hematologic diseases provide evidence that MSCs are indeed efficacious, but the significant heterogeneity in therapeutic approaches between studies raises new questions. The purpose of this review is to stimulate new preclinical and clinical trials to investigate these factors. First, we discuss recent clinical trials for pediatric diseases studying MSCs obtained from bone marrow, umbilical cord and umbilical cord blood, placenta, amniotic fluid, and adipose tissue. We then identify factors, some unique to pediatrics, which must be examined to optimize therapeutic efficacy, including route of administration, dose, timing of administration, the role of ex vivo differentiation, cell culture techniques, donor factors, host factors, and the immunologic implications of allogeneic therapy. Finally, we discuss some of the practicalities of bringing cell-based therapy into the clinic, including regulatory and manufacturing considerations. The aim of this review is to inform future studies seeking to maximize therapeutic efficacy for each disease and for each patient. Stem Cells Translational Medicine 2017;6:539-565.
Collapse
Affiliation(s)
- Christopher R. Nitkin
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Tracey L. Bonfield
- Division of Pulmonology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
42
|
Cytokines TNF-α, IL-6, IL-17F, and IL-4 Differentially Affect Osteogenic Differentiation of Human Adipose Stem Cells. Stem Cells Int 2016; 2016:1318256. [PMID: 27667999 PMCID: PMC5030432 DOI: 10.1155/2016/1318256] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022] Open
Abstract
During the initial stages of bone repair, proinflammatory cytokines are released within the injury site, quickly followed by a shift to anti-inflammatory cytokines. The effect of pro- and anti-inflammatory cytokines on osteogenic differentiation of mesenchymal stem cells is controversial. Here, we investigated the effect of the proinflammatory cytokines TNF-α, IL-6, IL-8, and IL-17F and the anti-inflammatory cytokine IL-4 on proliferation and osteogenic differentiation of human adipose stem cells (hASCs). hASCs were treated with TNF-α, IL-6, IL-8, IL-17F, or IL-4 (10 ng/mL) for 72 h mimicking bone repair. TNF-α reduced collagen type I gene expression but increased hASC proliferation and ALP activity. IL-6 also strongly enhanced ALP activity (18-fold), as well as bone nodule formation by hASCs. IL-8 did not affect proliferation or osteogenic gene expression but reduced bone nodule formation. IL-17F decreased hASC proliferation but enhanced ALP activity. IL-4 enhanced osteocalcin gene expression and ALP activity but reduced RUNX2 gene expression and bone nodule formation. In conclusion, all cytokines studied have both enhancing and reducing effects on osteogenic differentiation of hASCs, even when applied for 72 h only. Some cytokines, specifically IL-6, may be suitable to induce osteogenic differentiation of mesenchymal stem cells as a strategy for enhancing bone repair.
Collapse
|
43
|
Wang WZ. Microcirculatory Response In Vivo on Local Intraarterial Infusion of Autogenic Adipose-derived Stem Cells or Stromal Vascular Fraction. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e1067. [PMID: 27757364 PMCID: PMC5055030 DOI: 10.1097/gox.0000000000001067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/12/2016] [Indexed: 01/25/2023]
Abstract
Both adipose-derived stem cells (ASCs) and stromal vascular fraction (SVF) have been demonstrated to have regenerative properties with therapeutic potential for numerous diseases through local or topical applications. However, it is unclear whether ASC or SVF can be delivered systemically through an intra-arterial infusion. The purpose of this study was to examine the microcirculatory response in vivo on local intraarterial infusion of autogenic ASCs or SVF in a vascular pedicle isolated rat cremaster microcirculation model. MATERIALS AND METHODS Fat tissue was surgically harvested from the flanks of male Sprague-Dawley rats (n = 12) and processed for SVF isolation. Some SVF samples were cultured for 24 hours for ASC purification. The autogenic SVF (1 × 105) cells (n = 6) or purified ASC (1 × 105) cells (n = 6) cells were infused into the microcirculation of cremaster muscle at a speed of 0.05 mL/min through the cannulation of femoral artery. As this is a vascular pedicle isolated preparation, the infused SVF or ASC cells went nowhere but the cremaster muscle. The video image of the microcirculation was monitored in real time during infusion. RESULTS Arteriole diameter was measured as A1 (100-160 µm), A2 (40-80 µm), and A3/A4 (10-30 µm). Capillary perfusion was quantified in 18 capillary fields of each muscle. There was a significant increase in the diameter of terminal arterioles (P = 0.049) and the capillary density (P = 0.02) after ASC intraarterial infusion. However, a significant cell aggregation, embolisms, and arterial obstruction were observed in the microcirculation in every case during SVF infusion. CONCLUSIONS Intraarterial infusion is an appropriate route for the delivery of autogenic ASCs, but not of SVF. SVF-induced microembolisms were the reason for narrowing or blocking the lumen of terminal arterioles, resulting in no flow in the corresponding capillaries.
Collapse
Affiliation(s)
- Wei Z Wang
- Division of Plastic Surgery, Department of Surgery, School of Medicine, University of Nevada, Las Vegas, Nev
| |
Collapse
|
44
|
Development of Synthetic and Natural Materials for Tissue Engineering Applications Using Adipose Stem Cells. Stem Cells Int 2016; 2016:5786257. [PMID: 26977158 PMCID: PMC4764745 DOI: 10.1155/2016/5786257] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/09/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose stem cells have prominent implications in tissue regeneration due to their abundance and relative ease of harvest from adipose tissue and their abilities to differentiate into mature cells of various tissue lineages and secrete various growth cytokines. Development of tissue engineering techniques in combination with various carrier scaffolds and adipose stem cells offers great potential in overcoming the existing limitations constraining classical approaches used in plastic and reconstructive surgery. However, as most tissue engineering techniques are new and highly experimental, there are still many practical challenges that must be overcome before laboratory research can lead to large-scale clinical applications. Tissue engineering is currently a growing field of medical research; in this review, we will discuss the progress in research on biomaterials and scaffolds for tissue engineering applications using adipose stem cells.
Collapse
|