1
|
Tchoukalova YD, Shah MK, Myers CE, Zhang N, Lott DG. Optimization of seeding cell density for differentiation of adipose-derived stem cells into epithelial-like cells on bioengineered composite scaffolds. Differentiation 2025; 143:100870. [PMID: 40414118 DOI: 10.1016/j.diff.2025.100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
This study investigates the biological factors influencing the epithelial differentiation of adipose-derived stem cells (ASC) to develop an engineered upper airway construct. One fraction of ASC was seeded onto a fibrin sealant (Tisseel) matrix encompassing an additional equal fraction of ASC that has been integrated into a porous polyethylene scaffold (Medpor®). Constructs with ASC seeded at total densities of 5 × 105, 1 × 106, 2.5 × 106, and 5 × 106 cells cm-2 were cultured under submerged conditions for 11 days to achieve partial epithelial differentiation (PD). To simulate post-transplantation exposure to air and interaction with host epithelial cells, PD constructs with ASC at 5 × 106 cells cm-2 were transitioned to air-liquid interface (ALI) conditions for additional 10 days (PD/ALI-21d) or 21 days (PD/ALI-32d). The latter cultures were either maintained alone or co-cultured with bronchial epithelial cells (PD/ALI-32d + BEAS). Gene expressions of mesenchymal and epithelial basal, secretory, and ciliated cell markers were assessed and validated via immunohistochemistry. ASC seeded at 5 × 106 cells cm-2 achieved the highest partial epithelial differentiation, supporting the use of this density for further experiments. In PD/ALI-21d, basal and secretory epithelial marker gene expression significantly increased, while ciliated cell markers remained unchanged. In PD/ALI-32d, expression of basal and goblet cell markers and several mesenchymal stem cell markers decreased, but co-culturing with BEAS maintained the levels of their expression. These results indicate that long-term ALI cultures cannot sustain terminal differentiation of ASC into secretory phenotypes without co-culture with primary epithelial cells. In conclusion, partially differentiated ASC on constructs maintain a stem cell phenotype and may promote differentiation into basal/secretory phenotypes, but not ciliated cells. Enhancing ciliogenesis and ensuring ASC commitment to the epithelial lineage, require modifications to the study design.
Collapse
Affiliation(s)
- Yourka D Tchoukalova
- Mayo Clinic Arizona, Head and Neck Regenerative Medicine Laboratory, Scottsdale, AZ, USA
| | - Manisha K Shah
- Mayo Clinic Arizona, Head and Neck Regenerative Medicine Laboratory, Scottsdale, AZ, USA
| | - Cheryl E Myers
- Mayo Clinic Arizona, Head and Neck Regenerative Medicine Laboratory, Scottsdale, AZ, USA
| | - Nan Zhang
- Mayo Clinic Arizona, Department of Quantitative Health Science Research, Scottsdale, AZ, USA
| | - David G Lott
- Mayo Clinic Arizona, Head and Neck Regenerative Medicine Laboratory, Scottsdale, AZ, USA; Mayo Clinic Arizona, Department of Otolaryngology - Head and Neck Surgery, Division of Laryngology, Phoenix, AZ, USA.
| |
Collapse
|
2
|
Trotzier C, Bellanger C, Abdessadeq H, Delannoy P, Mojallal A, Auxenfans C. Deciphering influence of donor age on adipose-derived stem cells: in vitro paracrine function and angiogenic potential. Sci Rep 2024; 14:27589. [PMID: 39528480 PMCID: PMC11555058 DOI: 10.1038/s41598-024-73875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND As fat grafting is commonly used as a filler, Adipose-derived stem/stromal cells (ASC) have been reported to be key player in retention rate. Paracrine and differentiation potential of those cells confer them strong pro-angiogenic capacities. However, a full characterization of the influence of aging on ASC has not been reported yet. Here we've investigated the effect of age on paracrine function, stemness and angiogenic potential of ASC. METHODS ASC were extracted from young and old adult donors. We assessed stromal vascular fraction cell populations repartition, ASC stemness potential, capability to differentiate into mesenchymal lineages as well as their secretome. Angiogenic potential was assessed using a sprouting assay, an indirect co-culture of ASC and dermal microvascular endothelial cells (EC). Total vascular sprout length was measured, and co-culture soluble factors were quantified. Pro-angiogenic factors alone or in combination as well as ASC-conditioned medium (CM) were added to EC to assess sprouting induction. RESULTS Decrease of endothelial cells yield and percentage is observed in cells extracted from adipose tissue of older patients, whereas ASC percentage increased with age. Clonogenic potential of ASC is stable with age. ASC can differentiate into adipocytes, chondrocytes and osteoblasts, and aging does not alter this potential. Among the 25 analytes quantified, high levels of pro-angiogenic factors were found, but none is significantly modulated with age. ASC induce a significantly longer vascular sprouts compared to fibroblasts, and no difference was found between young and old ASC donors on that parameter. Higher concentrations of FGF-2, G-CSF, HGF and IL-8, and lower concentrations of VEGF-C were quantified in EC/ASC co-cultures compared to EC/fibroblasts co-cultures. EC/ASC from young donors secrete higher levels of VEGF-A compared to old ones. Neither soluble factor nor CM without cells are able to induce organized sprouts, highlighting the requirement of cell communication for sprouting. CM produced by ASC supporting development of long vascular sprouts promote sprouting in co-cultures that establish shorter sprouts. CONCLUSION Our results show cells from young and old donors exhibit no difference in all assessed parameters, suggesting all patients could be included in clinical applications. We emphasized the leading role of ASC in angiogenesis, without impairment with age, where secretome is a key but not sufficient actor.
Collapse
Affiliation(s)
- Chloe Trotzier
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France.
| | - Clement Bellanger
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Hakima Abdessadeq
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Philippe Delannoy
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Ali Mojallal
- Department of Plastic, Reconstructive and Aesthetic Surgery, La Croix Rousse Hospital, Bernard Lyon 1 University, Lyon, France
| | - Celine Auxenfans
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
3
|
Getova VE, Pinheiro-Machado E, Harmsen MC, Burgess JK, Smink AM. The role of extracellular matrix hydrogels and adipose-derived stromal cells in soft tissue vascularization - A systematic review. BIOMATERIALS ADVANCES 2024; 164:213986. [PMID: 39151272 DOI: 10.1016/j.bioadv.2024.213986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/12/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
Decellularized extracellular matrix (dECM) hydrogels loaded with adipose-derived stromal cells (ASC) or their conditioned medium (ASC CM) present a promising and versatile treatment approach for tissue vascularization and regeneration. These hydrogels are easy to produce, store, personalize, manipulate, and deliver to the target tissue. This literature review aimed to investigate the applications of dECM hydrogels with ASC or ASC CM for in vivo tissue vascularization. Fourteen experimental studies have been reviewed using vessel density as the primary outcome parameter for in vivo vascularization. The studies consistently reported an increased efficacy in augmenting angiogenesis by the ASC or ASC CM-loaded hydrogels compared to untreated controls. However, this systematic review shows the need to standardize procedures and characterization, particularly of the final administered product(s). The findings from these experimental studies highlight the potential of dECM hydrogel with ASC or ASC CM in novel tissue regeneration and regenerative medicine applications.
Collapse
Affiliation(s)
- Vasilena E Getova
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands
| | - Erika Pinheiro-Machado
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Martin C Harmsen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Alexandra M Smink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| |
Collapse
|
4
|
Lorentz KL, Marini AX, Bruk LA, Gupta P, Mandal BB, DiLeo MV, Weinbaum JS, Little SR, Vorp DA. Mesenchymal Stem Cell-Conditioned Media-Loaded Microparticles Enhance Acute Patency in Silk-Based Vascular Grafts. Bioengineering (Basel) 2024; 11:947. [PMID: 39329689 PMCID: PMC11428691 DOI: 10.3390/bioengineering11090947] [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: 06/28/2024] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Coronary artery disease leads to over 360,000 deaths annually in the United States, and off-the-shelf bypass graft options are currently limited and/or have high failure rates. Tissue-engineered vascular grafts (TEVGs) present an attractive option, though the promising mesenchymal stem cell (MSC)-based implants face uncertain regulatory pathways. In this study, "artificial MSCs" (ArtMSCs) were fabricated by encapsulating MSC-conditioned media (CM) in poly(lactic-co-glycolic acid) microparticles. ArtMSCs and control microparticles (Blank-MPs) were incubated over 7 days to assess the release of total protein and the vascular endothelial growth factor (VEGF-A); releasates were also assessed for cytotoxicity and promotion of smooth muscle cell (SMC) proliferation. Each MP type was loaded in previously published "lyogel" silk scaffolds and implanted as interposition grafts in Lewis rats for 1 or 8 weeks. Explanted grafts were assessed for patency and cell content. ArtMSCs had a burst release of protein and VEGF-A. CM increased proliferation in SMCs, but not after encapsulation. TEVG explants after 1 week had significantly higher patency rates with ArtMSCs compared to Blank-MPs, but similar to unseeded lyogel grafts. ArtMSC explants had lower numbers of infiltrating macrophages compared to Blank-MP explants, suggesting a modulation of inflammatory response by the ArtMSCs. TEVG explants after 8 weeks showed no significant difference in patency among the three groups. The ArtMSC explants showed higher numbers of SMCs and endothelial cells within the neotissue layer of the graft compared to Blank-MP explants. In sum, while the ArtMSCs had positive effects acutely, efficacy was lost in the longer term; therefore, further optimization is needed.
Collapse
Affiliation(s)
- Katherine L Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ande X Marini
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liza A Bruk
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Prerak Gupta
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Morgan V DiLeo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, PA 15261, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee Women's Research Institute, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
Vuorenpää H, Valtonen J, Penttinen K, Koskimäki S, Hovinen E, Ahola A, Gering C, Parraga J, Kelloniemi M, Hyttinen J, Kellomäki M, Aalto-Setälä K, Miettinen S, Pekkanen-Mattila M. Gellan gum-gelatin based cardiac models support formation of cellular networks and functional cardiomyocytes. Cytotechnology 2024; 76:483-502. [PMID: 38933872 PMCID: PMC11196475 DOI: 10.1007/s10616-024-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/06/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases remain as the most common cause of death worldwide. To reveal the underlying mechanisms in varying cardiovascular diseases, in vitro models with cells and supportive biomaterial can be designed to recapitulate the essential components of human heart. In this study, we analyzed whether 3D co-culture of cardiomyocytes (CM) with vascular network and with adipose tissue-derived mesenchymal stem/stromal cells (ASC) can support CM functionality. CM were cultured with either endothelial cells (EC) and ASC or with only ASC in hydrazide-modified gelatin and oxidized gellan gum hybrid hydrogel to form cardiovascular multiculture and myocardial co-culture, respectively. We studied functional characteristics of CM in two different cellular set-ups and analyzed vascular network formation, cellular morphology and orientation. The results showed that gellan gum-gelatin hydrogel supports formation of two different cellular networks and functional CM. We detected formation of a modest vascular network in cardiovascular multiculture and extensive ASC-derived alpha smooth muscle actin -positive cellular network in multi- and co-culture. iPSC-CM showed elongated morphology, partly aligned orientation with the formed networks and presented normal calcium transients, beating rates, and contraction and relaxation behavior in both setups. These 3D cardiac models provide promising platforms to study (patho) physiological mechanisms of cardiovascular diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00630-5.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Joona Valtonen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi Penttinen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Koskimäki
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emma Hovinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ahola
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Christine Gering
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jenny Parraga
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kellomäki
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katriina Aalto-Setälä
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Hospital, Tampere University Hospital, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
6
|
Arderiu G, Bejar MT, Civit-Urgell A, Peña E, Badimon L. Crosstalk of human coronary perivascular adipose-derived stem cells with vascular cells: role of tissue factor. Basic Res Cardiol 2024; 119:291-307. [PMID: 38430261 DOI: 10.1007/s00395-024-01037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
The coronary perivascular adipose tissue (cPVAT) has been associated to the burden of cardiovascular risk factors and to the underlying vessel atherosclerotic plaque severity. Although the "outside to inside" hypothesis of PVAT-derived-adipokine regulation of vessel function is currently accepted, whether the resident mesenchymal stem cells (ASCs) in PVAT have a regulatory role on the underlying vascular arterial smooth muscle cells (VSMCs) is not known. Here, we investigated the interactions between resident PVAT-ASCs and VSMCs. ASCs were obtained from PVAT overlying the left anterior descending (LAD) coronary artery of hearts removed at heart transplant operations. PVAT was obtained both from patients with non-ischemic and ischemic heart disease as the cause of heart transplant. ASCs were isolated from PVAT, phenotypically characterized by flow cytometry, functionally tested for proliferation, and differentiation. Crosstalk between ASCs and VSMCs was investigated by co-culture studies. ASCs were detected in the adventitia of the LAD-PVAT showing differentiation capacity and angiogenic potential. ASCs obtained from PVAT of non-ischemic and ischemic hearts showed different tissue factor (TF) expression levels, different VSMCs recruitment capacity through the axis ERK1/2-ETS1 signaling and different angiogenic potential. Induced upregulation of TF in ASCs isolated from ischemic PVAT rescued their angiogenic capacity in subcutaneously implanted plugs in mice, whereas silencing TF in ASCs decreased the proangiogenic capacity of non-ischemic ASCs. The results indicate for the first time a novel mechanism of regulation of VSMCs by PVAT-ASCs in angiogenesis, mediated by TF expression in ASCs. Regulation of TF in ASCs may become a therapeutic intervention to increase cardiac protection.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain.
- Ciber CV, Instituto Carlos III, Madrid, Spain.
| | - Maria Teresa Bejar
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Anna Civit-Urgell
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Barcelona, Spain
| | - Esther Peña
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Xiong Z, Hu Y, Jiang M, Liu B, Jin W, Chen H, Yang L, Han X. Hypoxic bone marrow mesenchymal stem cell exosomes promote angiogenesis and enhance endometrial injury repair through the miR-424-5p-mediated DLL4/Notch signaling pathway. PeerJ 2024; 12:e16953. [PMID: 38406291 PMCID: PMC10894593 DOI: 10.7717/peerj.16953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/25/2024] [Indexed: 02/27/2024] Open
Abstract
Background Currently, bone marrow mesenchymal stem cells (BMSCs) have been reported to promote endometrial regeneration in rat models of mechanically injury-induced uterine adhesions (IUAs), but the therapeutic effects and mechanisms of hypoxic BMSC-derived exosomes on IUAs have not been elucidated. Objective To investigate the potential mechanism by which the BMSCS-derived exosomal miR-424-5p regulates IUA angiogenesis through the DLL4/Notch signaling pathway under hypoxic conditions and promotes endometrial injury repair. Methods The morphology of the exosomes was observed via transmission electron microscopy, and the expression of exosome markers (CD9, CD63, CD81, and HSP70) was detected via flow cytometry and Western blotting. The expression of angiogenesis-related genes (Ang1, Flk1, Vash1, and TSP1) was detected via RT‒qPCR, and the expression of DLL4/Notch signaling pathway-related proteins (DLL4, Notch1, and Notch2) was detected via Western blotting. Cell proliferation was detected by a CCK-8 assay, and angiogenesis was assessed via an angiogenesis assay. The expression of CD3 was detected by immunofluorescence. The endometrial lesions of IUA rats were observed via HE staining, and the expression of CD3 and VEGFA was detected via immunohistochemistry. Results Compared with those in exosomes from normoxic conditions, miR-424-5p was more highly expressed in the exosomes from hypoxic BMSCs. Compared with those in normoxic BMSC-derived exosomes, the proliferation and angiogenesis of HUVECs were significantly enhanced after treatment with hypoxic BMSC-derived exosomes, and these effects were weakened after inhibition of miR-424-5p. miR-424-5p can target and negatively regulate the expression of DLL4, promote the expression of the proangiogenic genes Ang1 and Flk1, and inhibit the expression of the antiangiogenic genes Vash1 and TSP1. The effect of miR-424-5p can be reversed by overexpression of DLL4. In IUA rats, treatment with hypoxic BMSC exosomes and the miR-424-5p mimic promoted angiogenesis and improved endometrial damage. Conclusion The hypoxic BMSC-derived exosomal miR-424-5p promoted angiogenesis and improved endometrial injury repair by regulating the DLL4/Notch signaling pathway, which provides a new idea for the treatment of IUAs.
Collapse
Affiliation(s)
- Zhenghua Xiong
- Department of Gynecology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- Department of Gynecology, Yan’an Hospital Affiliated to Kunming Medical University/Yan’an Hospital of Kunming City, Kunming, Yunnan, China
| | - Yong Hu
- Department of Gynecology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Min Jiang
- Department of Gynecology, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Beibei Liu
- Department of Gynecology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenjiao Jin
- Department of Gynecology, Yan’an Hospital Affiliated to Kunming Medical University/Yan’an Hospital of Kunming City, Kunming, Yunnan, China
| | - Huiqin Chen
- Department of Gynecology, Chuxiong Hospital of Traditional Chinese Medicine, Chuxiong, Yunnan, China
| | - Linjuan Yang
- Department of Gynecology and Obstetrics, Baoshan Hospital of Traditional Chinese Medicine, Baoshan, Yunnan, China
| | - Xuesong Han
- Department of Gynecology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- Department of Gynecology, Yan’an Hospital Affiliated to Kunming Medical University/Yan’an Hospital of Kunming City, Kunming, Yunnan, China
| |
Collapse
|
8
|
Deng S, Zhu F, Dai K, Wang J, Liu C. Harvest of functional mesenchymal stem cells derived from in vivo osteo-organoids. BIOMATERIALS TRANSLATIONAL 2023; 4:270-279. [PMID: 38282704 PMCID: PMC10817801 DOI: 10.12336/biomatertransl.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/14/2023] [Accepted: 11/30/2023] [Indexed: 01/30/2024]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) play a crucial role in stem cell therapy and are extensively used in regenerative medicine research. However, current methods for harvesting BM-MSCs present challenges, including a low yield of primary cells, long time of in vitro expansion, and diminished differentiation capability after passaging. Meanwhile mesenchymal stem cells (MSCs) recovered from cell banks also face issues like toxic effects of cryopreservation media. In this study, we provide a detailed protocol for the isolation and evaluation of MSCs derived from in vivo osteo-organoids, presenting an alternative to autologous MSCs. We used recombinant human bone morphogenetic protein 2-loaded gelatin sponge scaffolds to construct in vivo osteo-organoids, which were stable sources of MSCs with large quantity, high purity, and strong stemness. Compared with protocols using bone marrow, our protocol can obtain large numbers of high-purity MSCs in a shorter time (6 days vs. 12 days for obtaining passage 1 MSCs) while maintaining higher stemness. Notably, we found that the in vivo osteo-organoid-derived MSCs exhibited stronger anti-replicative senescence capacity during passage and amplification, compared to BM-MSCs. The use of osteo-organoid-derived MSCs addresses the conflict between the limitations of autologous cells and the risks associated with allogeneic sources in stem cell transplantation. Consequently, our protocol emerges as a superior alternative for both stem cell research and tissue engineering.
Collapse
Affiliation(s)
- Shunshu Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Fuwei Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
- Shanghai University, Shanghai, China
| |
Collapse
|
9
|
Mankuzhy P, Dharmarajan A, Perumalsamy LR, Sharun K, Samji P, Dilley RJ. The role of Wnt signaling in mesenchymal stromal cell-driven angiogenesis. Tissue Cell 2023; 85:102240. [PMID: 37879288 DOI: 10.1016/j.tice.2023.102240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023]
Abstract
Development, growth, and remodeling of blood vessels occur through an intricate process involving cell differentiation, proliferation, and rearrangement by cell migration under the direction of various signaling pathways. Recent reports highlight that resident and exogenous mesenchymal stromal cells (MSCs) have the potential to regulate the neovascularization process through paracrine secretion of proangiogenic factors. Recent research has established that the vasculogenic potential of MSCs is regulated by several signaling pathways, including the Wnt signaling pathway, and their interplay. These findings emphasize the complex nature of the vasculogenic process and underscore the importance of understanding the underlying molecular mechanisms for the development of effective cell-based therapies in regenerative medicine. This review provides an updated briefing on the canonical and non-canonical Wnt signaling pathways and summarizes the recent reports of both in vitro and in vivo studies with the involvement of MSCs of various sources in the vasculogenic process mediated by Wnt signaling pathways. Here we outline the current understanding of the plausible role of the Wnt signaling pathway, specifically in MSC-regulated angiogenesis.
Collapse
Affiliation(s)
- Pratheesh Mankuzhy
- Department of Surgery and Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, 6009 Perth, Australia; College of Veterinary and Animal Sciences - Mannuthy, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India.
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India; School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Perth, Western Australia, Australia; School of Human Sciences, Faculty of Life Sciences, University of Western Australia, 6009 Perth, Australia
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Priyanka Samji
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Rodney J Dilley
- Department of Surgery and Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, 6009 Perth, Australia
| |
Collapse
|
10
|
Tsimponis A, Dionyssiou D, Papamitsou T, Demiri E. The effect of host tissue and radiation on fat-graft survival: A comparative experimental study. JPRAS Open 2023; 38:134-146. [PMID: 37929062 PMCID: PMC10623108 DOI: 10.1016/j.jpra.2023.08.009] [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: 06/28/2023] [Accepted: 08/27/2023] [Indexed: 11/07/2023] Open
Abstract
Because lipofilling is often associated with various reconstructive procedures, especially breast reconstructions, improving fat-graft retention remains a major concern for plastic surgeons. We conducted an experimental protocol in a rat model simulating an autologous breast reconstruction method using the fat-augmented latissimus dorsi myocutaneous (LDM) flap. This study aimed to compare the survival rates of autologous adipocytes when injected subcutaneously and intramuscularly and to evaluate the role of recipient host tissue, volume of the injected fat, and postoperative radiation on fat-graft retention. Thirty rats were divided into five groups (A, B, C, D, and E), of six rats each. All animals underwent a pedicled LDM flap transfer to the anterior thoracic wall, and different volumes of autologous fat were injected into three recipient areas, namely, the pectoralis major and latissimus dorsi muscles and the subcutaneous tissue of the flap's skin island, as follows: 1 mL of fat was injected in total in group A, 2 mL in groups B and D, and 5 mL in group C. Group D animals received postoperative radiation (24 Gy), whereas group E animals (controls) did not undergo any fat grafting procedure. Eight weeks after surgery, adipocyte survival was assessed in all groups using histological and immunochemistry techniques. The results showed that the pectoralis major muscle was the substrate with the highest adipocyte survival rates, which were proportional to the amount of fat injected, followed by the latissimus dorsi muscle and the subcutaneous tissue. Increased volumes of transplanted fat into the subcutaneous tissue did not correspond to increased adipocyte survival. Irradiation of host tissues resulted in a statistically significant decrease in surviving adipocytes in all three recipient sites (p<0.001). Our study strongly suggests that muscle ensures optimal fat-graft retention, whereas postoperative radiation negatively affects adipocyte survival following fat transplantation.
Collapse
Affiliation(s)
- Antonios Tsimponis
- Department of Plastic Surgery, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki Greece
| | - Dimitrios Dionyssiou
- Department of Plastic Surgery, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki Greece
| | - Theodora Papamitsou
- Laboratory of Histology-Embryology, Aristotle University of Thessaloniki, Greece
| | - Efterpi Demiri
- Department of Plastic Surgery, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki Greece
| |
Collapse
|
11
|
Sharma D, Sharma A, Hu L, Chen TA, Voon S, Bayless KJ, Goldman J, Walsh AJ, Zhao F. Perfusability and immunogenicity of implantable pre-vascularized tissues recapitulating features of native capillary network. Bioact Mater 2023; 30:184-199. [PMID: 37589031 PMCID: PMC10425689 DOI: 10.1016/j.bioactmat.2023.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
Vascularization is a key pre-requisite to engineered anatomical scale three dimensional (3-D) constructs to ensure their nutrient and oxygen supply upon implantation. Presently, engineered pre-vascularized 3-D tissues are limited to only micro-scale hydrogels, which meet neither the anatomical scale needs nor the complexity of natural extracellular matrix (ECM) environments. Anatomical scale perfusable constructs are critically needed for translational applications. To overcome this challenge, we previously developed pre-vascularized ECM sheets with long and oriented dense microvascular networks. The present study further evaluated the patency, perfusability and innate immune response toward these pre-vascularized constructs. Macrophage-co-cultured pre-vascularized constructs were evaluated in vitro to confirm micro-vessel patency and perturbations in macrophage metabolism. Subcutaneously implanted pre-vascularized constructs remained viable and formed a functional anastomosis with host vasculature within 3 days of implantation. This completely biological pre-vascularized construct holds great potential as a building block to engineer perfusable anatomical scale tissues.
Collapse
Affiliation(s)
- Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Archita Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Linghao Hu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Sarah Voon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Kayla J. Bayless
- School of Medicine, Texas A&M University, College Station, TX, United States
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, United States
| | - Alex J. Walsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
12
|
Suominen S, Hyypijev T, Venäläinen M, Yrjänäinen A, Vuorenpää H, Lehti-Polojärvi M, Räsänen M, Seppänen A, Hyttinen J, Miettinen S, Aalto-Setälä K, Viiri LE. Improvements in Maturity and Stability of 3D iPSC-Derived Hepatocyte-like Cell Cultures. Cells 2023; 12:2368. [PMID: 37830581 PMCID: PMC10571736 DOI: 10.3390/cells12192368] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology enables differentiation of human hepatocytes or hepatocyte-like cells (iPSC-HLCs). Advances in 3D culturing platforms enable the development of more in vivo-like liver models that recapitulate the complex liver architecture and functionality better than traditional 2D monocultures. Moreover, within the liver, non-parenchymal cells (NPCs) are critically involved in the regulation and maintenance of hepatocyte metabolic function. Thus, models combining 3D culture and co-culturing of various cell types potentially create more functional in vitro liver models than 2D monocultures. Here, we report the establishment of 3D cultures of iPSC-HLCs alone and in co-culture with human umbilical vein endothelial cells (HUVECs) and adipose tissue-derived mesenchymal stem/stromal cells (hASCs). The 3D cultures were performed as spheroids or on microfluidic chips utilizing various biomaterials. Our results show that both 3D spheroid and on-chip culture enhance the expression of mature liver marker genes and proteins compared to 2D. Among the spheroid models, we saw the best functionality in iPSC-HLC monoculture spheroids. On the contrary, in the chip system, the multilineage model outperformed the monoculture chip model. Additionally, the optical projection tomography (OPT) and electrical impedance tomography (EIT) system revealed changes in spheroid size and electrical conductivity during spheroid culture, suggesting changes in cell-cell connections. Altogether, the present study demonstrates that iPSC-HLCs can successfully be cultured in 3D as spheroids and on microfluidic chips, and co-culturing iPSC-HLCs with NPCs enhances their functionality. These 3D in vitro liver systems are promising human-derived platforms usable in various liver-related studies, specifically when using patient-specific iPSCs.
Collapse
Affiliation(s)
- Siiri Suominen
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Tinja Hyypijev
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Mari Venäläinen
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Mari Lehti-Polojärvi
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Mikko Räsänen
- Department of Technical Physics, University of Eastern Finland, 70210 Kuopio, Finland
| | - Aku Seppänen
- Department of Technical Physics, University of Eastern Finland, 70210 Kuopio, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Katriina Aalto-Setälä
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
- Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland
| | - Leena E. Viiri
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| |
Collapse
|
13
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [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: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
14
|
Lynn JV, Lalchandani KB, Daniel M, Urlaub KM, Ettinger RE, Nelson NS, Donneys A, Buchman SR. Adipose-Derived Stem Cells Enhance Graft Incorporation and Mineralization in a Murine Model of Irradiated Mandibular Nonvascularized Bone Grafting. Ann Plast Surg 2023; 91:154-158. [PMID: 37450875 DOI: 10.1097/sap.0000000000003598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
BACKGROUND Nonvascularized bone grafting represents a practical method of mandibular reconstruction. However, the destructive effects of radiotherapy on native bone preclude the use of nonvascularized bone grafts in head and neck cancer patients. Adipose-derived stem cells have been shown to enhance bone healing and regeneration in numerous experimental models. The purpose of this study was to determine the impact of adipose-derived stem cells on nonvascularized bone graft incorporation in a murine model of irradiated mandibular reconstruction. METHODS Thirty isogenic rats were randomly divided into 3 groups: nonvascularized bone graft (control), radiation with nonvascularized bone graft (XRT), and radiation with nonvascularized bone graft and adipose-derived stem cells (ASC). Excluding the control group, all rats received a human-equivalent dose of radiation. All groups underwent mandibular reconstruction of a critical-sized defect with a nonvascularized bone graft from the contralateral hemimandible. After a 60-day recovery period, graft incorporation and bone mineralization were compared between groups. RESULTS Compared with the control group, the XRT group demonstrated significantly decreased graft incorporation (P = 0.011), bone mineral density (P = 0.005), and bone volume fraction (P = 0.001). Compared with the XRT group, the ASC group achieved a significantly increased graft incorporation (P = 0.006), bone mineral density (P = 0.005), and bone volume fraction (P = 0.013). No significant differences were identified between the control and ASC groups. CONCLUSIONS Adipose-derived stem cells enhance nonvascularized bone graft incorporation in the setting of human-equivalent radiation.
Collapse
Affiliation(s)
- Jeremy V Lynn
- From the Craniofacial Research Laboratory, University of Michigan, Ann Arbor, MI
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
SUMMARY Over the past 30 years, there has been a dramatic increase in the use of autologous fat grafting for soft-tissue augmentation and to improve facial skin quality. Several studies have highlighted the impact of aging on adipose tissue, leading to a decrease of adipose tissue volume and preadipocyte proliferation and increase of fibrosis. Recently, there has been a rising interest in adipose tissue components, including adipose-derived stem/stromal cells (ASCs) because of their regenerative potential, including inflammation, fibrosis, and vascularization modulation. Because of their differentiation potential and paracrine function, ASCs have been largely used for fat grafting procedures, as they are described to be a key component in fat graft survival. However, many parameters as surgical procedures or adipose tissue biology could change clinical outcomes. Variation on fat grafting methods have led to numerous inconsistent clinical outcomes. Donor-to-donor variation could also be imputed to ASCs, tissue inflammatory state, or tissue origin. In this review, the authors aim to analyze (1) the parameters involved in graft survival, and (2) the effect of aging on adipose tissue components, especially ASCs, that could lead to a decrease of skin regeneration and fat graft retention. CLINICAL RELEVANCE STATEMENT This review aims to enlighten surgeons about known parameters that could play a role in fat graft survival. ASCs and their potential mechanism of action in regenerative medicine are more specifically described.
Collapse
|
16
|
Zhang Y, Lv P, Li Y, Zhang Y, Cheng C, Hao H, Yue H. Inflammatory Cytokine Interleukin-6 (IL-6) Promotes the Proangiogenic Ability of Adipose Stem Cells from Obese Subjects via the IL-6 Signaling Pathway. Curr Stem Cell Res Ther 2023; 18:93-104. [PMID: 36883256 DOI: 10.2174/1574888x17666220429103935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/05/2022] [Accepted: 03/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The prevalence of obesity, as well as obesity-induced chronic inflammatory diseases, is increasing worldwide. Chronic inflammation is related to the complex process of angiogenesis, and we found that adipose-derived stem cells from obese subjects (obADSCs) had proangiogenic features, including higher expression levels of interleukin-6 (IL-6), Notch ligands and receptors, and proangiogenic cytokines, than those from control subjects. We hypothesized that IL-6 and Notch signaling pathways are essential for regulating the proangiogenic characteristics of obADSCs. OBJECTIVE This study aimed to investigate whether the inflammatory cytokine interleukin 6 (IL-6) promotes the proangiogenic capacity of adipose stem cells in obese subjects via the IL-6 signaling pathway. METHODS We compared the phenotype analysis as well as cell doubling time, proliferation, migration, differentiation, and proangiogenic properties of ADSCs in vitro. Moreover, we used small interfering RNAs to inhibit the gene and protein expression of IL-6. RESULTS We found that ADSCs isolated from control individuals (chADSCs) and obADSCs had similar phenotypes and growth characteristics, and chADSCs had a stronger differentiation ability than obADSCs. However, obADSCs were more potent in promoting EA.hy926 cell migration and tube formation than chADSCs in vitro. We confirmed that IL-6 siRNA significantly reduced the transcriptional level of IL-6 in obADSCs, thereby reducing the expression of vascular endothelial growth factor (VEGF)- A, VEGF receptor 2, transforming growth factor β, and Notch ligands and receptors in obADSCs. CONCLUSION The finding suggests that inflammatory cytokine interleukin-6 (IL-6) promotes the proangiogenic ability of obADSCs via the IL-6 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Pengju Lv
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Yalong Li
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Yonghui Zhang
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Chaofei Cheng
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Hongbo Hao
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center, City University of New York, New York, 10031, USA
| | - Han Yue
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
17
|
Brembilla NC, Vuagnat H, Boehncke WH, Krause KH, Preynat-Seauve O. Adipose-Derived Stromal Cells for Chronic Wounds: Scientific Evidence and Roadmap Toward Clinical Practice. Stem Cells Transl Med 2022; 12:17-25. [PMID: 36571216 PMCID: PMC9887085 DOI: 10.1093/stcltm/szac081] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/16/2022] [Indexed: 12/27/2022] Open
Abstract
Chronic wounds, ie, non-healing ulcers, have a prevalence of ~1% in the general population. Chronic wounds strongly affect the quality of life and generate considerable medical costs. A fraction of chronic wounds will heal within months of appropriate treatment; however, a significant fraction of patients will develop therapy-refractory chronic wounds, leading to chronic pain, infection, and amputation. Given the paucity of therapeutic options for refractory wounds, cell therapy and in particular the use of adipose-derived stromal cells (ASC) has emerged as a promising concept. ASC can be used as autologous or allogeneic cells. They can be delivered in suspension or in 3D cultures within scaffolds. ASC can be used without further processing (stromal vascular fraction of the adipose tissue) or can be expanded in vitro. ASC-derived non-cellular components, such as conditioned media or exosomes, have also been investigated. Many in vitro and preclinical studies in animals have demonstrated the ASC efficacy on wounds. ASC efficiency appears to occurs mainly through their regenerative secretome. Hitherto, the majority of clinical trials focused mainly on safety issues. However more recently, a small number of randomized, well-controlled trials provided first convincing evidences for a clinical efficacy of ASC-based chronic wound therapies in humans. This brief review summarizes the current knowledge on the mechanism of action, delivery and efficacy of ASC in chronic wound therapy. It also discusses the scientific and pharmaceutical challenges to be solved before ASC-based wound therapy enters clinical reality.
Collapse
Affiliation(s)
- Nicolo C Brembilla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland
| | - Hubert Vuagnat
- Program for Wounds and Wound Healing, Care Directorate, Geneva University Hospitals, Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Laboratory of Therapy and Stem Cells, Geneva University Hospitals, Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Corresponding author: Olivier Preynat-Seauve, PATIM, 1 rue Michel Servet CH-1211 Geneva 4, Switzerland. Tel: +41223794139;
| |
Collapse
|
18
|
Almalki SG. Adipose-derived mesenchymal stem cells and wound healing: Potential clinical applications in wound repair. Saudi Med J 2022; 43:1075-1086. [PMID: 36261194 PMCID: PMC9994497 DOI: 10.15537/smj.2022.43.10.20220522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023] Open
Abstract
Delayed and chronic wounds result from the dysregulation of molecular and cellular events associated with wound healing, including migration, inflammation, angiogenesis, extracellular matrix (ECM) remodeling, and re-epithelialization. Adipose tissue is an abundant, easily accessible, and rich source of mesenchymal stem cells (MSCs) with high therapeutic potential. In addition to their capability to differentiate into various lineages with specialized functions, adipose-derived MSCs (AMSCs) can mediate to the wound repair process through the secretion of different growth factors and mediators rather than making structural contribution alone. Adipose-derived MSCs mediate the formation of blood vessels, recruit progenitor cells, stimulate cell differentiation and ECM formation, and promote wound healing by releasing immune mediators and exosomes. Herein, we discuss and review the therapeutic potential of AMSCs for wound repair via acceleration of wound closure, re-epithelialization, enhancement of angiogenesis and immunomodulation of prolonged inflammatory responses, as well as the current challenges in clinical implementation.
Collapse
Affiliation(s)
- Sami G. Almalki
- From the Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| |
Collapse
|
19
|
Angiogenic Potential of Co-Cultured Human Umbilical Vein Endothelial Cells and Adipose Stromal Cells in Customizable 3D Engineered Collagen Sheets. J Funct Biomater 2022; 13:jfb13030107. [PMID: 35997445 PMCID: PMC9397038 DOI: 10.3390/jfb13030107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 01/25/2023] Open
Abstract
The wound healing process is much more complex than just the four phases of hemostasis, inflammation, proliferation, and maturation. Three-dimensional (3D) scaffolds made of biopolymers or ECM molecules using bioprinting can be used to promote the wound healing process, especially for complex 3D tissue lesions like chronic wounds. Here, a 3D-printed mold has been designed to produce customizable collagen type-I sheets containing human umbilical vein endothelial cells (HUVECs) and adipose stromal cells (ASCs) for the first time. In these 3D collagen sheets, the cellular activity leads to a restructuring of the collagen matrix. The upregulation of the growth factors Serpin E1 and TIMP-1 could be demonstrated in the 3D scaffolds with ACSs and HUVECs in co-culture. Both growth factors play a key role in the wound healing process. The capillary-like tube formation of HUVECs treated with supernatant from the collagen sheets revealed the secretion of angiogenic growth factors. Altogether, this demonstrates that collagen type I combined with the co-cultivation of HUVECs and ACSs has the potential to accelerate the process of angiogenesis and, thereby, might promote wound healing.
Collapse
|
20
|
Mousavi A, Stefanek E, Jafari A, Ajji Z, Naghieh S, Akbari M, Savoji H. Tissue-engineered heart chambers as a platform technology for drug discovery and disease modeling. BIOMATERIALS ADVANCES 2022; 138:212916. [PMID: 35913255 DOI: 10.1016/j.bioadv.2022.212916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Current drug screening approaches are incapable of fully detecting and characterizing drug effectiveness and toxicity of human cardiomyocytes. The pharmaceutical industry uses mathematical models, cell lines, and in vivo models. Many promising drugs are abandoned early in development, and some cardiotoxic drugs reach humans leading to drug recalls. Therefore, there is an unmet need to have more reliable and predictive tools for drug discovery and screening applications. Biofabrication of functional cardiac tissues holds great promise for developing a faithful 3D in vitro disease model, optimizing drug screening efficiencies enabling precision medicine. Different fabrication techniques including molding, pull spinning and 3D bioprinting were used to develop tissue-engineered heart chambers. The big challenge is to effectively organize cells into tissue with structural and physiological features resembling native tissues. Some advancements have been made in engineering miniaturized heart chambers that resemble a living pump for drug screening and disease modeling applications. Here, we review the currently developed tissue-engineered heart chambers and discuss challenges and prospects.
Collapse
Affiliation(s)
- Ali Mousavi
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Evan Stefanek
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Zineb Ajji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Mohsen Akbari
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada; Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada.
| |
Collapse
|
21
|
Asimakopoulos D, Anastasatos JM. Cell-Assisted Lipotransfer in Breast Augmentation Surgery: Clinical Outcomes and Considerations for Future Research. Cureus 2022; 14:e22763. [PMID: 35371878 PMCID: PMC8971120 DOI: 10.7759/cureus.22763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 11/30/2022] Open
Abstract
Autologous fat transfer is a widely used surgical technique, chosen by numerous plastic surgeons for breast augmentation surgery. This technique is based on three steps: 1. harvesting of the lipoaspirate from the patient, 2. centrifugation and removal of the top, oily, layer, and 3. implantation in the patient’s breast(s). It has been associated with various complications, including post-surgical fat resorption, as measured quantitatively with MRI, CT, and other 3D-quantification systems. Adipose-derived stem cells have been explored as a means of addressing fat resorption. They can be separated from the lipoaspirate following centrifugation, and enzymatically purified from unwanted debris, with collagenase, forming the stromal vascular fraction. The stromal vascular fraction is then recombined with the graft volume prior to implantation. This novel technique, referred to as “cell-assisted lipotransfer”, has shown promising results in terms of reducing fat resorption. These results are due to the pro-angiogenic and pro-adipogenic ability of the stem cells, which allow the graft to address the conditions of ischemia more effectively than autologous fat transfer. The aim of this review is to explore the ways in which cell-assisted lipotransfer is different from the autologous fat transfer, as well as how and why adipose-derived stem cells may contribute towards limiting fat resorption. The immunological background of these cells is discussed in detail, while grounds for further development are discussed, by means of the administration of external growth factors, which could, potentially, maximize outcomes, while limiting complications.
Collapse
|
22
|
In Vivo Evaluation of Mechanically Processed Stromal Vascular Fraction in a Chamber Vascularized by an Arteriovenous Shunt. Pharmaceutics 2022; 14:pharmaceutics14020417. [PMID: 35214149 PMCID: PMC8880586 DOI: 10.3390/pharmaceutics14020417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a promising source for regenerative purposes. To study the in vivo fate of the mSVF, we herein used a vascularized tissue engineering chamber that insulates the target mSVF from the surrounding environment. In contrast to previous models, we propose an arteriovenous (AV) shunt between saphenous vessels in rats without a venous graft. Mechanical SVF was processed from the fat pads of male Sprague Dawley rats, mixed with a fibrin hydrogel and implanted into an inguinal tissue engineering chamber. An arteriovenous shunt was established between saphenous artery and vein. On the contralateral side, an mSVF-fibrin hydrogel mix without vascular axis served as a non-vascularized control. After two and six weeks, rats were sacrificed for further analysis. Mechanical SVF showed significant numbers of mesenchymal stromal cells. Vascularized mSVF explants gained weight over time. Perilipin and CD31 expression were significantly higher in the mSVF explants after six weeks while no difference in DAPI positive cells, collagen deposition and FABP4 expression was observed. Morphologically, no differentiated adipocytes but a dense cell-rich tissue with perilipin-positive cells was found after six weeks. The phosphorylation of ERK1/2 was significantly enhanced after six weeks while Akt activation remained unaltered. Finally, mSVF explants stably expressed and released VEGF, bFGF and TGFb. Vascularized mSVF is able to proliferate and express adipocyte-specific markers. The AV shunt model is a valuable refinement of currently existing AV loop models in the rat which contributes to the fundamental 3R principles of animal research.
Collapse
|
23
|
Shadmani A, Razmkhah M, Jalalpoor MH, Lari SY, Eghtedari M. Autologous Activated Omental versus Allogeneic Adipose Tissue-Derived Mesenchymal Stem Cells in Corneal Alkaline Injury: An Experimental Study. J Curr Ophthalmol 2021; 33:136-142. [PMID: 34409223 PMCID: PMC8365576 DOI: 10.4103/joco.joco_246_20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 01/23/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose: To compare the effects of two types of mesenchymal stem cells (MSCs), activated omental cells (AOCs), and adipose tissue-derived stem cells (ADSCs) in the healing process of animal model of ocular surface alkali injury. Methods: An alkaline burn was induced on the ocular surfaces of eighteen rats divided randomly into three groups. The first and second groups received subconjunctival AOCs and ADSCs, respectively. The control group received normal saline subconjunctival injection. On the 90th day after the injury, the eyes were examined using slit-lamp biomicroscopy. Corneal neovascularization and scarring were graded in a masked fashion. Histological evaluation of the corneal scar was performed, and the number of inflammatory cells was evaluated. Results: Corneal neovascularization scores revealed higher neovascularization in the control (0.49 ± 0.12) than the AOC (0.80 ± 0.20, P = 0.01) and ADSC groups (0.84 ± 0.24, P = 0.007). There were no statistically significant differences between the neovascularization score of the AOC and ADSC groups (P > 0.05). According to histologic evaluation, stromal infiltration was significantly more in the control group compared to AOC and ADSC groups (P < 0.05). Conclusions: Our results suggest that MSCs, even with different sources, can be used to promote wound healing after corneal chemical burns. However, the ease of harvesting ADSC from more superficial fat sources makes this method more clinically applicable.
Collapse
Affiliation(s)
- Athar Shadmani
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Masoomeh Eghtedari
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Ophthalmology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
24
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
25
|
Später T, Ampofo E, Menger MD, Laschke MW. Combining Vascularization Strategies in Tissue Engineering: The Faster Road to Success? Front Bioeng Biotechnol 2020; 8:592095. [PMID: 33364230 PMCID: PMC7752995 DOI: 10.3389/fbioe.2020.592095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/20/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- Thomas Später
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
26
|
Human mesenchymal stromal/stem cells recruit resident pericytes and induce blood vessels maturation to repair experimental spinal cord injury in rats. Sci Rep 2020; 10:19604. [PMID: 33177535 PMCID: PMC7658254 DOI: 10.1038/s41598-020-76290-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is considered to mediate the beneficial effects of mesenchymal cell therapy in spinal cord injury. After a moderate balloon-compression injury in rats, injections of either human adipose tissue-derived stromal/stem cells (hADSCs) or their conditioned culture media (CM-hADSC) elicited angiogenesis around the lesion site. Both therapies increased vascular density, but the presence of hADSCs in the tissue was required for the full maturation of new blood vessels. Only animals that received hADSC significantly improved their open field locomotion, assessed by the BBB score. Animals that received CM-hADSC only, presented haemorrhagic areas and lack pericytes. Proteomic analyses of human angiogenesis-related factors produced by hADSCs showed that both pro- and anti-angiogenic factors were produced by hADSCs in vitro, but only those related to vessel maturation were detectable in vivo. hADSCs produced PDGF-AA only after insertion into the injured spinal cord. hADSCs attracted resident pericytes expressing NG2, α-SMA, PDGF-Rβ and nestin to the lesion, potentially contributing to blood vessel maturation. We conclude that the presence of hADSCs in the injured spinal cord is essential for tissue repair.
Collapse
|
27
|
Kiaie N, Gorabi AM, Ahmadi Tafti SH, Rabbani S. Pre-vascularization Approaches for Heart Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00172-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
28
|
Robbins ER, Pins GD, Laflamme MA, Gaudette GR. Creation of a contractile biomaterial from a decellularized spinach leaf without ECM protein coating: An in vitro study. J Biomed Mater Res A 2020; 108:2123-2132. [PMID: 32323417 PMCID: PMC7725356 DOI: 10.1002/jbm.a.36971] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 01/08/2023]
Abstract
Myocardial infarction (MI) results in the death of cardiac tissue, decreases regional contraction, and can lead to heart failure. Tissue engineered cardiac patches containing human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) can restore contractile function. However, cells within thick patches require vasculature for blood flow. Recently, we demonstrated fibronectin coated decellularized leaves provide a suitable scaffold for hiPS-CMs. Yet, the necessity of this additional coating step is unclear. Therefore, we compared hiPS-CM behavior on decellularized leaves coated with collagen IV or fibronectin extracellular matrix (ECM) proteins to noncoated leaves for up to 21 days. Successful coating was verified by immunofluorescence. Similar numbers of hiPS-CMs adhered to coated and noncoated decellularized leaves for 21 days. At Day 14, collagen IV coated leaves contracted more than noncoated leaves (3.25 ± 0.39% vs. 1.54 ± 0.60%; p < .05). However, no differences in contraction were found between coated leaves, coated tissue culture plastic (TCP), noncoated leaves, or noncoated TCP at other time points. No significant differences were observed in hiPS-CM spreading or sarcomere lengths on leaves with or without coating. This study demonstrates that cardiac scaffolds can be created from decellularized leaves without ECM coatings. Noncoated decellularized leaf surfaces facilitate robust cell attachment for an engineered tissue patch.
Collapse
Affiliation(s)
- Emily R. Robbins
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - George D. Pins
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, Ontario, Canada
| | - Glenn R. Gaudette
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| |
Collapse
|
29
|
Nellinger S, Schmidt I, Heine S, Volz A, Kluger PJ. Adipose stem cell‐derived extracellular matrix represents a promising biomaterial by inducing spontaneous formation of prevascular‐like structures by mvECs. Biotechnol Bioeng 2020; 117:3160-3172. [DOI: 10.1002/bit.27481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/24/2020] [Accepted: 06/24/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Isabelle Schmidt
- School of Applied Chemistry Reutlingen University Reutlingen Germany
| | - Simon Heine
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Ann‐Cathrin Volz
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Petra J. Kluger
- School of Applied Chemistry Reutlingen University Reutlingen Germany
| |
Collapse
|
30
|
Shukla L, Yuan Y, Shayan R, Greening DW, Karnezis T. Fat Therapeutics: The Clinical Capacity of Adipose-Derived Stem Cells and Exosomes for Human Disease and Tissue Regeneration. Front Pharmacol 2020; 11:158. [PMID: 32194404 PMCID: PMC7062679 DOI: 10.3389/fphar.2020.00158] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Fat grafting is a well-established surgical technique used in plastic surgery to restore deficient tissue, and more recently, for its putative regenerative properties. Despite more frequent use of fat grafting, however, a scientific understanding of the mechanisms underlying either survival or remedial benefits of grafted fat remain lacking. Clinical use of fat grafts for breast reconstruction in tissues damaged by radiotherapy first provided clues regarding the clinical potential of stem cells to drive tissue regeneration. Healthy fat introduced into irradiated tissues appeared to reverse radiation injury (fibrosis, scarring, contracture and pain) clinically; a phenomenon since validated in several animal studies. In the quest to explain and enhance these therapeutic effects, adipose-derived stem cells (ADSCs) were suggested as playing a key role and techniques to enrich ADSCs in fat, in turn, followed. Stem cells - the body's rapid response 'road repair crew' - are on standby to combat tissue insults. ADSCs may exert influences either by releasing paracrine-signalling factors alone or as cell-free extracellular vesicles (EVs, exosomes). Alternatively, ADSCs may augment vital immune/inflammatory processes; or themselves differentiate into mature adipose cells to provide the 'building-blocks' for engineered tissue. Regardless, adipose tissue constitutes an ideal source for mesenchymal stem cells for therapeutic application, due to ease of harvest and processing; and a relative abundance of adipose tissue in most patients. Here, we review the clinical applications of fat grafting, ADSC-enhanced fat graft, fat stem cell therapy; and the latest evolution of EVs and nanoparticles in healing, cancer and neurodegenerative and multiorgan disease.
Collapse
Affiliation(s)
- Lipi Shukla
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Yinan Yuan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| | - Ramin Shayan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia.,Plastic, Hand and Faciomaxillary Surgery Unit, Alfred Hospital, Prahran, VIC, Australia.,Department of Plastic and Reconstructive Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Tara Karnezis
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| |
Collapse
|
31
|
Debels H, Palmer J, Han XL, Poon C, Abberton K, Morrison W. In vivo tissue engineering of an adipose tissue flap using fat grafts and Adipogel. J Tissue Eng Regen Med 2020; 14:633-644. [PMID: 32090506 DOI: 10.1002/term.3027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 01/27/2020] [Accepted: 02/04/2020] [Indexed: 12/17/2022]
Abstract
For decades, plastic surgeons have spent considerable effort exploring anatomical regions for free flap design. More recently, tissue-engineering approaches have been utilised in an attempt to grow transplantable tissue flaps in vivo. The aim of this study was to engineer a fat flap with a vascular pedicle by combining autologous fat grafts and a novel acellular hydrogel (Adipogel) in an established tissue-engineering model comprising a chamber and blood vessel loop. An arteriovenous loop was created in the rat groin from the femoral vessels and positioned inside a perforated polycarbonate chamber. In Group 1, the chamber contained minced, centrifuged autologous fat; in Group 2, Adipogel was added to the graft; and in Group 3, Adipogel alone was used. Constructs were histologically examined at 6 and 12 weeks. In all groups, new tissue was generated. Adipocytes, although appearing viable in the graft at the time of insertion, were predominantly nonviable at 6 weeks. However, by 12 weeks, new fat had formed in all groups and was significantly greater in the combined fat/Adipogel group. No significant difference was seen in final construct total volume or construct neovascularisation between the groups. This study demonstrated that a pedicled adipose flap can be generated in rats by combining a blood vessel loop, an adipogenic hydrogel, and a lipoaspirate equivalent. Success appears to be based on adipogenesis rather than on adipocyte survival, and consistent with our previous work, this adipogenesis occurred subsequent to graft death and remodelling. The regenerative process was significantly enhanced in the presence of Adipogel.
Collapse
Affiliation(s)
- Heidi Debels
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,Department of Plastic and Reconstructive Surgery, Free University Brussels (VUB), Belgium.,Department of Plastic Surgery, Maastricht University, Maastricht, The Netherlands
| | - Jason Palmer
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Xiao-Lian Han
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia
| | - Christopher Poon
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Keren Abberton
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Wayne Morrison
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| |
Collapse
|
32
|
Sparks DS, Savi FM, Saifzadeh S, Schuetz MA, Wagels M, Hutmacher DW. Convergence of Scaffold-Guided Bone Reconstruction and Surgical Vascularization Strategies-A Quest for Regenerative Matching Axial Vascularization. Front Bioeng Biotechnol 2020; 7:448. [PMID: 31998712 PMCID: PMC6967032 DOI: 10.3389/fbioe.2019.00448] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
The prevalent challenge facing tissue engineering today is the lack of adequate vascularization to support the growth, function, and viability of tissue engineered constructs (TECs) that require blood vessel supply. The research and clinical community rely on the increasing knowledge of angiogenic and vasculogenic processes to stimulate a clinically-relevant vascular network formation within TECs. The regenerative matching axial vascularization approach presented in this manuscript incorporates the advantages of flap-based techniques for neo-vascularization yet also harnesses the in vivo bioreactor principle in a more directed "like for like" approach to further assist regeneration of the specific tissue type that is lost, such as a corticoperiosteal flap in critical sized bone defect reconstruction.
Collapse
Affiliation(s)
- David S Sparks
- Centre for Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Department of Plastic & Reconstructive Surgery, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Southside Clinical Division, School of Medicine, University of Queensland, Woolloongabba, QLD, Australia
| | - Flavia Medeiros Savi
- Centre for Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Siamak Saifzadeh
- Centre for Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Medical Engineering Research Facility, Queensland University of Technology, Chermside, QLD, Australia
| | - Michael A Schuetz
- Department of Orthopaedic Surgery, Royal Brisbane Hospital, Herston, QLD, Australia.,Jamieson Trauma Institute, Royal Brisbane Hospital, Herston, QLD, Australia
| | - Michael Wagels
- Department of Plastic & Reconstructive Surgery, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Southside Clinical Division, School of Medicine, University of Queensland, Woolloongabba, QLD, Australia.,Australian Centre for Complex Integrated Surgical Solutions, Woolloongabba, QLD, Australia
| | - Dietmar W Hutmacher
- Centre for Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,ARC Centre for Additive Bio-Manufacturing, Queensland University of Technology, Kelvin Grove, QLD, Australia
| |
Collapse
|
33
|
The Role of Adipose-Derived Stem Cells, Dermal Regenerative Templates, and Platelet-Rich Plasma in Tissue Engineering-Based Treatments of Chronic Skin Wounds. Stem Cells Int 2020; 2020:7056261. [PMID: 32399048 PMCID: PMC7199611 DOI: 10.1155/2020/7056261] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
The continuous improvements in the field of both regenerative medicine and tissue engineering have allowed the design of new and more efficacious strategies for the treatment of chronic or hard-to-heal skin wounds, which represent heavy burden, from a medical and economic point of view. These novel approaches are based on the usage of three key methodologies: stem cells, growth factors, and biomimetic scaffolds. These days, the adipose tissue can be considered the main source of multipotent mesenchymal stem cells, especially adipose-derived stem cells (ASCs). ASCs are easily accessible from various fat depots and show an intrinsic plasticity in giving rise to cell types involved in wound healing and angiogenesis. ASCs can be found in fat grafts, historically used in the treatment of chronic wounds, and have been evaluated as such in both animal models and human trials, to exploit their capability of accelerating wound closure and inducing a correct remodeling of the newly formed fibrovascular tissue. Since survival and fitness of ASCs need to be improved, they are now employed in conjunction with advanced wound dressings, together with dermal regenerative templates and platelet-rich plasma (as a source of growth and healing factors). In this work, we provide an overview of the current knowledge on the topic, based on existing studies and on our own experience.
Collapse
|
34
|
Ejaz A, Greenberger JS, Rubin PJ. Understanding the mechanism of radiation induced fibrosis and therapy options. Pharmacol Ther 2019; 204:107399. [DOI: 10.1016/j.pharmthera.2019.107399] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
|
35
|
Wang Z, Mithieux SM, Weiss AS. Fabrication Techniques for Vascular and Vascularized Tissue Engineering. Adv Healthc Mater 2019; 8:e1900742. [PMID: 31402593 DOI: 10.1002/adhm.201900742] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Impaired or damaged blood vessels can occur at all levels in the hierarchy of vascular systems from large vasculatures such as arteries and veins to meso- and microvasculatures such as arterioles, venules, and capillary networks. Vascular tissue engineering has become a promising approach for fabricating small-diameter vascular grafts for occlusive arteries. Vascularized tissue engineering aims to fabricate meso- and microvasculatures for the prevascularization of engineered tissues and organs. The ideal small-diameter vascular graft is biocompatible, bridgeable, and mechanically robust to maintain patency while promoting tissue remodeling. The desirable fabricated meso- and microvasculatures should rapidly integrate with the host blood vessels and allow nutrient and waste exchange throughout the construct after implantation. A number of techniques used, including engineering-based and cell-based approaches, to fabricate these synthetic vasculatures are herein explored, as well as the techniques developed to fabricate hierarchical structures that comprise multiple levels of vasculature.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Suzanne M. Mithieux
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
- Bosch Institute University of Sydney NSW 2006 Australia
- Sydney Nano Institute University of Sydney NSW 2006 Australia
| |
Collapse
|
36
|
Electrical stimulation promotes the angiogenic potential of adipose-derived stem cells. Sci Rep 2019; 9:12076. [PMID: 31427631 PMCID: PMC6700204 DOI: 10.1038/s41598-019-48369-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
Autologous fat transfer (AFT) is limited by post-operative volume loss due to ischemia-induced cell death in the fat graft. Previous studies have demonstrated that electrical stimulation (ES) promotes angiogenesis in a variety of tissues and cell types. In this study we investigated the effects of ES on the angiogenic potential of adipose-derived stem cells (ASC), important progenitor cells in fat grafts with proven angiogenic potential. Cultured human ASC were electrically stimulated for 72 hours after which the medium of stimulated (ES) and non-stimulated (control) ASC was analysed for angiogenesis-related proteins by protein array and ELISA. The functional effect of ES on angiogenesis was then assessed in vitro and in vivo. Nine angiogenesis-related proteins were detected in the medium of electrically (non-)stimulated ASC and were quantified by ELISA. The pro-angiogenic proteins VEGF and MCP-1 were significantly increased following ES compared to controls, while the anti-angiogenic factor Serpin E1/PAI-1 was significantly decreased. Despite increased levels of anti-angiogenic TSP-1 and TIMP-1, medium of ES-treated ASC significantly increased vessel density, total vessel network length and branching points in chorio-allantoic membrane assays. In conclusion, our proof-of-concept study showed that ES increased the angiogenic potential of ASC both in vitro and in vivo.
Collapse
|
37
|
Huang B, Huang LF, Zhao L, Zeng Z, Wang X, Cao D, Yang L, Ye Z, Chen X, Liu B, He TC, Wang X. Microvesicles (MIVs) secreted from adipose-derived stem cells (ADSCs) contain multiple microRNAs and promote the migration and invasion of endothelial cells. Genes Dis 2019; 7:225-234. [PMID: 32215292 PMCID: PMC7083715 DOI: 10.1016/j.gendis.2019.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) such as microvesicles (MIVs) play an important role in intercellular communications. MIVs are small membrane vesicles sized 100–1000 nm in diameter that are released by many types of cells, such as mesenchymal stem cells (MSCs), tumor cells and adipose-derived stem cells (ADSC). As EVs can carry out autocrine and paracrine functions by controlling multiple cell processes, it is conceivable that EVs can be used as delivery vehicles for treating several clinical conditions, such as to improve cardiac angiogenesis after myocardial infarction (MI). Here, we seek to investigate whether ADSC-derived MIVs contain microRNAs that regulate angiogenesis and affect cell migration of endothelial cells. We first characterized the ADSC-derived MIVs and found that the MIVs had a size range of 100–300 nm, and expressed the MIV marker protein Alix. We then analyzed the microRNAs in ADSCs and ADSC-derived MIVs and demonstrated that ADSC-derived MIVs selectively released a panel of microRNAs, several of which were related to angiogenesis, including two members of the let-7 family. Furthermore, we demonstrated that ADSC-derived MIVs promoted the cell migration and invasion of the HUVEC endothelial cells. The PKH26-labeled ADSC-derived MIVs were effectively uptaken into the cytoplasm of HUVEC cells. Collectively, our results demonstrate that the ADSC-derived MIVs can promote migration and invasion abilities of endothelial cells, suggesting pro-angiogenetic potential. Future studies should focus on investigating the roles and mechanisms through which ADSC-derived MIVs regulate angiogenesis.
Collapse
Affiliation(s)
- Bo Huang
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Lin-Feng Huang
- Department of Clinical Laboratory Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, China
| | - Ling Zhao
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Zongyue Zeng
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Xi Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Daigui Cao
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of Orthopaedic Surgery, Chongqing General Hospital Affiliated with the University of Chinese Academy of Sciences, Chongqing, 400013, China
| | - Lijuan Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Zhenyu Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xian Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of Clinical Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266061, China
| | - Bin Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.,Department of Biology, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Xiaozhong Wang
- Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| |
Collapse
|
38
|
Kenar H, Ozdogan CY, Dumlu C, Doger E, Kose GT, Hasirci V. Microfibrous scaffolds from poly(l-lactide-co-ε-caprolactone) blended with xeno-free collagen/hyaluronic acid for improvement of vascularization in tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:31-44. [PMID: 30678916 DOI: 10.1016/j.msec.2018.12.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/03/2018] [Accepted: 12/05/2018] [Indexed: 02/08/2023]
Abstract
Success of 3D tissue substitutes in clinical applications depends on the presence of vascular networks in their structure. Accordingly, research in tissue engineering is focused on the stimulation of angiogenesis or generation of a vascular network in the scaffolds prior to implantation. A novel, xeno-free, collagen/hyaluronic acid-based poly(l-lactide-co-ε-caprolactone) (PLC/COL/HA) (20/9.5/0.5 w/w/w) microfibrous scaffold was produced by electrospinning. Collagen types I and III, and hyaluronic acid were isolated from human umbilical cords and blended with the GMP grade PLC. When compared with PLC scaffolds the PLC/COL/HA had higher water uptake capacity (103% vs 66%) which may have contributed to the decrease in its Young's Modulus (from 1.31 to 0.89 MPa). The PLC/COL/HA better supported adipose tissue-derived mesenchymal stem cell (AT MSC) adhesion; within 24 h the cell number on the PLC/COL/HA scaffolds was 3 fold higher. Co-culture of human umbilical vein endothelial cells and AT MSCs induced capillary formation on both scaffold types, but the PLC/COL/HA led to formation of interconnected vessels whose total length was 1.6 fold of the total vessel length on PLC. Clinical use of this scaffold would eliminate the immune response triggered by xenogeneic collagen and transmission of animal-borne diseases while promoting a better vascular network formation.
Collapse
Affiliation(s)
- Halime Kenar
- Experimental and Clinical Research Center, Diabetes and Obesity Research Laboratory, Kocaeli University, Turkey; Polymer Science and Technology Dept., Graduate School of Natural and Applied Sciences, Kocaeli University, Turkey; BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey.
| | - Candan Yilmaz Ozdogan
- Experimental and Clinical Research Center, Diabetes and Obesity Research Laboratory, Kocaeli University, Turkey; Department of Biology, Graduate School of Natural and Applied Sciences, Kocaeli University, Turkey
| | - Cansu Dumlu
- Polymer Science and Technology Dept., Graduate School of Natural and Applied Sciences, Kocaeli University, Turkey
| | - Emek Doger
- Department of Gynecology and Obstetrics, Kocaeli University, Turkey
| | - Gamze Torun Kose
- BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey; Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Vasif Hasirci
- BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey; Department of Biological Sciences, Middle East Technical University (METU), Ankara, Turkey
| |
Collapse
|
39
|
Novel non-angiogenic role for mesenchymal stem cell-derived vascular endothelial growth factor on keratinocytes during wound healing. Cytokine Growth Factor Rev 2018; 44:69-79. [PMID: 30470511 DOI: 10.1016/j.cytogfr.2018.11.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 12/21/2022]
Abstract
With chronic wounds remaining a substantial healthcare issue, new therapies are sought to improve patient outcomes. Various studies have explored the benefits of promoting angiogenesis in wounds by targeting proangiogenic factors such as Vascular Endothelial Growth Factor (VEGF) family members to improve wound healing. Along similar lines, Mesenchymal Stem Cell (MSC) secretions, usually containing VEGF, have been used to improve angiogenesis in wound healing via a paracrine mechanism. Recent evidence for keratinocyte VEGF receptor expression, as well as proliferative and chemotactic responses by keratinocytes to exogenous VEGFA in vitro implies distinct non-angiogenic actions for VEGF during wound healing. In this review, we discuss the expression of VEGF family members and their receptors in keratinocytes in relation to the potential for wound healing treatments. We also explore recent findings of MSC secreted paracrine wound healing activity on keratinocytes. We report here the concept of keratinocyte wound healing responses driven by MSC-derived VEGF that is supported in the literature, providing a new mechanism for cell-free therapy of chronic wounds.
Collapse
|
40
|
Kiaie N, Aghdam RM, Tafti SHA, Gorabi AM. Stem Cell-Mediated Angiogenesis in Tissue Engineering Constructs. Curr Stem Cell Res Ther 2018; 14:249-258. [PMID: 30394215 DOI: 10.2174/1574888x13666181105145144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/09/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022]
Abstract
Angiogenesis has always been a concern in the field of tissue engineering. Poor vascularization of engineered constructs is a problem for the clinical success of these structures. Among the various methods employed to induce angiogenesis, stem cells provide a promising tool for the future. The present review aims to present the application of stem cells in the induction of angiogenesis. Additionally, it summarizes recent advancements in stem cell-mediated angiogenesis of different tissue engineering constructs.
Collapse
Affiliation(s)
- Nasim Kiaie
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.,Department of Tissue Engineering, Amirkabir University of Technology, Tehran 15875, Iran
| | - Rouhollah M Aghdam
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed H Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita M Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Yap KK, Yeoh GC, Morrison WA, Mitchell GM. The Vascularised Chamber as an In Vivo Bioreactor. Trends Biotechnol 2018; 36:1011-1024. [DOI: 10.1016/j.tibtech.2018.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
|
42
|
Resveratrol improves human umbilical cord-derived mesenchymal stem cells repair for cisplatin-induced acute kidney injury. Cell Death Dis 2018; 9:965. [PMID: 30237401 PMCID: PMC6148224 DOI: 10.1038/s41419-018-0959-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/06/2018] [Accepted: 08/01/2018] [Indexed: 12/29/2022]
Abstract
Human umbilical cord-derived mesenchymal stem cells (hucMSCs) are a promising tool for damaged tissues repair, especially for the kidney. However, their efficacy requires improvement. In order to optimize the clinical utility of hucMSCs, we adopted a strategy of treating hucMSCs with 20 μmol/L of resveratrol (Res-hucMSCs), applying it in a cisplatin-induced acute kidney injury model. Interestingly, we found that Res-hucMSCs exhibited a more efficient repairing effect than did hucMSCs. Resveratrol-promoted hucMSCs secreted platelet-derived growth factor-DD (PDGF-DD) into renal tubular cells resulting in downstream phosphorylation of extracellular signal-regulated kinase (ERK), which inhibited renal tubular cells apoptosis. In contrast, PDGF-DD knockdown impaired the renal protection of Res-hucMSCs. In addition, angiogenesis induced by PDGF-DD in endothelial cells was also involved in the renal protection of Res-hucMSCs. The conditioned medium of Res-hucMSCs accelerated proliferation and migration of vascular endothelial cells in vitro and CD31 was in a high-level expression in Res-hucMSCs group in vivo. Nevertheless, the angiogenesis was abrogated when Res-hucMSCs were treated with PDGF-DD siRNA. In conclusion, our findings showed that resveratrol-modified hucMSCs activated ERK pathway in renal tubular cells and promoted angiogenesis in endothelial cells via paracrine PDGF-DD, which could be a novel strategy for enhancing the therapy efficacy of hucMSCs in cisplatin-induced kidney injury.
Collapse
|
43
|
Tanaka Y, Hamamoto Y, Niyazi A, Nagasao T, Ueno M, Tabata Y. Effects of platelet-rich plasma on tissue-engineered vascularized flaps in an in vivo chamber. J Plast Reconstr Aesthet Surg 2018; 71:1062-1068. [DOI: 10.1016/j.bjps.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 02/06/2018] [Accepted: 02/17/2018] [Indexed: 11/30/2022]
|
44
|
Fakoya AOJ, Otohinoyi DA, Yusuf J. Current Trends in Biomaterial Utilization for Cardiopulmonary System Regeneration. Stem Cells Int 2018; 2018:3123961. [PMID: 29853910 PMCID: PMC5949153 DOI: 10.1155/2018/3123961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/15/2017] [Accepted: 03/01/2018] [Indexed: 12/28/2022] Open
Abstract
The cardiopulmonary system is made up of the heart and the lungs, with the core function of one complementing the other. The unimpeded and optimal cycling of blood between these two systems is pivotal to the overall function of the entire human body. Although the function of the cardiopulmonary system appears uncomplicated, the tissues that make up this system are undoubtedly complex. Hence, damage to this system is undesirable as its capacity to self-regenerate is quite limited. The surge in the incidence and prevalence of cardiopulmonary diseases has reached a critical state for a top-notch response as it currently tops the mortality table. Several therapies currently being utilized can only sustain chronically ailing patients for a short period while they are awaiting a possible transplant, which is also not devoid of complications. Regenerative therapeutic techniques now appear to be a potential approach to solve this conundrum posed by these poorly self-regenerating tissues. Stem cell therapy alone appears not to be sufficient to provide the desired tissue regeneration and hence the drive for biomaterials that can support its transplantation and translation, providing not only physical support to seeded cells but also chemical and physiological cues to the cells to facilitate tissue regeneration. The cardiac and pulmonary systems, although literarily seen as just being functionally and spatially cooperative, as shown by their diverse and dissimilar adult cellular and tissue composition has been proven to share some common embryological codevelopment. However, necessitating their consideration for separate review is the immense adult architectural difference in these systems. This review also looks at details on new biological and synthetic biomaterials, tissue engineering, nanotechnology, and organ decellularization for cardiopulmonary regenerative therapies.
Collapse
Affiliation(s)
| | | | - Joshua Yusuf
- All Saints University School of Medicine, Roseau, Dominica
- All Saints University School of Medicine, Kingstown, Saint Vincent and the Grenadines
| |
Collapse
|
45
|
Wang G, Zheng Y, Wang Y, Cai Z, Liao N, Liu J, Zhang W. Co-culture system of hepatocytes and endothelial cells: two in vitro approaches for enhancing liver-specific functions of hepatocytes. Cytotechnology 2018; 70:1279-1290. [PMID: 29675734 DOI: 10.1007/s10616-018-0219-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 04/09/2018] [Indexed: 01/16/2023] Open
Abstract
Although hepatocyte transplantation and bioartificial liver support system provide new promising opportunities for those patients waiting for liver transplantation, hepatocytes are easily losing liver-specific functions by using the common in vitro cultured methods. The co-culture strategies with mimicking the in vivo microenvironment would facilitate the maintenance of liver-specific functions of hepatocytes. Considering that hepatocytes and endothelial cells (ECs) account for 80-90% of total cell populations in the liver, hepatocytes and ECs were directly co-cultured with hepatic stellate cells (HSCs) or adipose tissue-derived stem cells (ADSCs) at a ratio of 700:150:3 or 14:3:3 in the present study, and the liver-specific functions were carefully analyzed. Our results showed that the two co-culture systems presented the enhanced liver-specific functions through promoting secretion of urea and ALB and increasing the expressions of ALB, CYP3A4 and HNF4α, and the vessel-like structure in the co-culture system consisted of hepatocytes, ECs and ADSCs. Hence, our results suggested that the directly co-culture of hepatocytes and ECs with HSCs or ADSCs could significantly improve liver-specific functions of hepatocytes, and the co-culture system could further promote angiogenesis of ECs at a later stage. Therefore, this study provides potential interesting in vitro strategies for enhancing liver-specific functions of hepatocytes.
Collapse
Affiliation(s)
- Gaoxiong Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China
| | - Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350007, People's Republic of China
| | - Wenmin Zhang
- Department of Pathology, Fujian Medical University, Fuzhou, 350004, People's Republic of China.
| |
Collapse
|
46
|
Siegel KR, Clevenger TN, Clegg DO, Proctor DA, Proctor CS. Adipose Stem Cells Incorporated in Fibrin Clot Modulate Expression of Growth Factors. Arthroscopy 2018; 34:581-591. [PMID: 29100775 DOI: 10.1016/j.arthro.2017.08.250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/30/2017] [Accepted: 08/03/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the platelet capture rate of whole blood fibrin clots and the expression, secretion, and retention of the growth factors vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF) from fibrin clots and to determine how these levels may be modulated by allogeneic adipose-derived stem cells (ASCs). METHODS Whole blood from 10 human volunteers was transferred to a clotting device and the platelet capture rate determined. Two experimental conditions and 1 control were evaluated over 2 weeks in vitro. Clots made from human whole blood without ASCs, clot(-)ASC, were compared with clots with ASCs incorporated, clot(+)ASC, and a control group of synthetic polyethylene glycol gels with ASCs incorporated, control(+)ASCs. All conditions were examined for secretion and retention of VEGF, PDGF, and bFGF via enzyme-linked immunosorbent assay and immunohistochemistry. The analysis of platelet retention for clots made with this device was performed. RESULTS Enzyme-linked immunosorbent assay analysis showed significantly higher (P < .001) secretion of VEGF in clot(+)ASC compared with clot(-)ASC or control(+)ASC. In contrast, clot(-)ASC produced soluble PDGF, and the addition of ASCs results in decreased soluble PDGF with concomitant increases in PDGF immunoreactivity of ASCs. Soluble bFGF levels were low in clot(-)ASC, and were found to increase at early time points in clot(+)ASC. Furthermore, bFGF immunoreactivity could be detected in clot(+)ASC, whereas no bFGF immunoreactivity is present in clot(-)ASC or control(+)ASC. Control(+)ASC displayed a spike in bFGF secretion at day 0, which may be due to a stress response elicited by the encapsulation process. Approximately 98% of available platelets in whole blood were concentrated in the clot on formation. CONCLUSIONS Fibrin clots made by this method retain high concentrations of platelets, and when incorporated with ASCs show modulated secretion and immunoreactivity of VEGF, PDGF, and bFGF. CLINICAL RELEVANCE Whole blood fibrin clots capture platelets and release growth factors, and the addition of ASCs increases VEGF release for up to 2 weeks after clot formation. This suggests that whole blood fibrin clots may be a viable scaffold and delivery vehicle for future stem cell treatments.
Collapse
Affiliation(s)
- Kelsy R Siegel
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, U.S.A..
| | - Tracy N Clevenger
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, U.S.A
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, U.S.A
| | - Duncan A Proctor
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, U.S.A
| | | |
Collapse
|
47
|
Abstract
Angiogenesis plays an important role not only in the growth and regeneration of tissues in humans but also in pathological conditions such as inflammation, degenerative disease and the formation of tumors. Angiogenesis is also vital in thick engineered tissues and constructs, such as those for the heart and bone, as these can face difficulties in successful implantation if they are insufficiently vascularized or unable to connect to the host vasculature. Considerable research has been carried out on angiogenic processes using a variety of approaches. Pathological angiogenesis has been analyzed at the cellular level through investigation of cell migration and interactions, modeling tissue level interactions between engineered blood vessels and whole organs, and elucidating signaling pathways involved in different angiogenic stimuli. Approaches to regenerative angiogenesis in ischemic tissues or wound repair focus on the vascularization of tissues, which can be broadly classified into two categories: scaffolds to direct and facilitate tissue growth and targeted delivery of genes, cells, growth factors or drugs that promote the regeneration. With technological advancement, models have been designed and fabricated to recapitulate the innate microenvironment. Moreover, engineered constructs provide not only a scaffold for tissue ingrowth but a reservoir of agents that can be controllably released for therapeutic purposes. This review summarizes the current approaches for modeling pathological and regenerative angiogenesis in the context of micro-/nanotechnology and seeks to bridge these two seemingly distant aspects of angiogenesis. The ultimate aim is to provide insights and advances from various models in the realm of angiogenesis studies that can be applied to clinical situations.
Collapse
Affiliation(s)
- Li-Jiun Chen
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan.
| | | |
Collapse
|
48
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
49
|
Starokozhko V, Groothuis GMM. Challenges on the road to a multicellular bioartificial liver. J Tissue Eng Regen Med 2017; 12:e227-e236. [PMID: 27943623 DOI: 10.1002/term.2385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/28/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Abstract
Over recent years, the progress in the development of a bioartificial liver (BAL) as an extracorporeal device or as a tissue engineered transplantable organ has been immense. However, many important BAL characteristics that are necessary to meet clinical demands have not been sufficiently addressed. This review describes the existing challenges in the development of a BAL for clinical applications, highlighting multicellularity and sinusoidal microarchitecture as crucial BAL characteristics to fulfil various liver functions. Currently available sources of nonparenchymal liver cells, such as endothelial cells, cholangiocytes and macrophages, used in BAL development are defined. Also, we discuss the recent studies on the reconstruction of the complex liver sinusoid microarchitecture using various liver cell types. Moreover, we highlight some other aspects of a BAL, such as liver zonation and formation of a vascular as well as biliary network for an adequate delivery, biotransformation and removal of substrates and waste products. Finally, the benefits of a multicellular BAL for the pharmaceutical industry are briefly described. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Viktoriia Starokozhko
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands
| | - Geny M M Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands
| |
Collapse
|
50
|
Flow-Induced Axial Vascularization: The Arteriovenous Loop in Angiogenesis and Tissue Engineering. Plast Reconstr Surg 2017; 138:825-835. [PMID: 27673517 DOI: 10.1097/prs.0000000000002554] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Fabrication of a viable vascular network providing oxygen supply is identified as one crucial limiting factor to generate more complex three-dimensional constructs. The arteriovenous loop model provides initial blood supply and has a high angioinductive potency, making it suitable for vascularization of larger, tissue-engineered constructs. Also because of its angiogenic capabilities the arteriovenous loop is recently also used as a model to evaluate angiogenesis in vivo. This review summarizes the history of the arteriovenous loop model in research and its technical and surgical aspects. Through modifications of the isolation chamber and its containing matrices, tissue generation can be enhanced. In addition, matrices can be used as release systems for local application of growth factors, such as vascular endothelial growth factor and basic fibroblast growth factor, to affect vascular network formation. A special focus in this review is set on the assessment of angiogenesis in the arteriovenous loop model. This model provides good conditions for assessment of angiogenesis with the initial cell-free environment of the isolation chamber, which is vascularized by the arteriovenous loop. Because of the angiogenic capabilities of the arteriovenous loop model, different attempts were performed to create functional tissue in the isolation chamber for potential clinical application. Arteriovenous loops in combination with autologous bone marrow aspirate were already used to reconstruct large bone defects in humans.
Collapse
|