1
|
Lai Z, Niu X, Chen X, Lu F, Zhang Y, Yuan Y. Composite Microparticles of Fat Graft and GFR Matrigel Improved Volume Retention by Promoting Cell Migration and Vessel Regeneration. Aesthetic Plast Surg 2024; 48:1993-2001. [PMID: 38302709 DOI: 10.1007/s00266-022-03145-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/09/2022] [Indexed: 02/03/2024]
Abstract
BACKGROUND The retention volume of autologous fat grafts decreases after transplantation due to limited nutrition infiltration and insufficient blood supply. Structural fat grafts and the 3M (multipoint, multitunnel, and multilayer) injection technique have been considered to improve the survival of grafts; however, it is difficult for surgeons to practice in the clinic because grafts tend to gather into a cluster, especially in large volume fat grafting. Therefore, we hypothesize that prefabricated microparticle fat grafts (PFMG) may improve the retention rate. METHODS The C57BL/6 mouse fat particles were embedded in growth factor-reduced (GFR)-Matrigel to detect cell migration by immunofluorescence staining in vitro. PFMG was prepared by mixing mouse fat particles and GFR Matrigel in a 1:1 volume ratio and injected subcutaneously into C57BL/6 mice. Fat particles mixed with PBS in equal volume served as control group. The grafts were harvested at 1, 4, 8, and 12 weeks after sacrifice. The retention rate of grafts at each time point was measured, and the structural alterations were detected by SEM. Fat necrosis and blood vessel density were evaluated by histological analysis. RESULTS CD34+ cells are migrated from the PFMG and formed a tree-like tubular network in the in vitro study. The retention rate was higher in the PFMG group than in the control group at week 12 (38% vs. 30%, p < 0.05). After transplantation, the dissociated structure of fat particles was maintained in PFMG by SEM analysis. Histological analysis of PFMG confirmed less fat necrosis and more blood vessel density in the PFMG group at the early stage than in the control group. The GFR Matrigel was displaced by adipose tissue with time. CONCLUSIONS In this study, we developed a novel fat grafting method, PFMG that dispersed fat grafts and maintained the structure after transplantation. High volume retention volume of PFMG was achieved by promoting cell migration and vessel regeneration. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Zhuhao Lai
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, 219 Moganshan Road, 310005, Hangzhou, People's Republic of China
| | - Xingtang Niu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xihang Chen
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yuchen Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yi Yuan
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Rasouli M, Fattahi R, Nuoroozi G, Zarei-Behjani Z, Yaghoobi M, Hajmohammadi Z, Hosseinzadeh S. The role of oxygen tension in cell fate and regenerative medicine: implications of hypoxia/hyperoxia and free radicals. Cell Tissue Bank 2024; 25:195-215. [PMID: 37365484 DOI: 10.1007/s10561-023-10099-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
Oxygen pressure plays an integral role in regulating various aspects of cellular biology. Cell metabolism, proliferation, morphology, senescence, metastasis, and angiogenesis are some instances that are affected by different tensions of oxygen. Hyperoxia or high oxygen concentration, enforces the production of reactive oxygen species (ROS) that disturbs physiological homeostasis, and consequently, in the absence of antioxidants, cells and tissues are directed to an undesired fate. On the other side, hypoxia or low oxygen concentration, impacts cell metabolism and fate strongly through inducing changes in the expression level of specific genes. Thus, understanding the precise mechanism and the extent of the implication of oxygen tension and ROS in biological events is crucial to maintaining the desired cell and tissue function for application in regenerative medicine strategies. Herein, a comprehensive literature review has been performed to find out the impacts of oxygen tensions on the various behaviors of cells or tissues.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Student Research Committee, Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Ghader Nuoroozi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Zeinab Zarei-Behjani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, Zanjan University, Zanjan, Iran
| | - Zeinab Hajmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran.
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Talaei-Khozani T, Yaghoubi A. An overview of post transplantation events of decellularized scaffolds. Transpl Immunol 2022; 74:101640. [PMID: 35667545 DOI: 10.1016/j.trim.2022.101640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/19/2022]
Abstract
Regenerative medicine and tissue engineering are reasonable techniques for repairing failed tissues and could be a suitable alternative to organ transplantation. One of the most widely used methods for preparing bioscaffolds is the decellularization procedure. Although cell debris and DNA are removed from the decellularized tissues, important compositions of the extracellular matrix including proteins, proteoglycans, and glycoproteins are nearly preserved. Moreover, the obtained scaffolds have a 3-dimensional (3D) structure, appropriate naïve mechanical properties, and good biocompatibility. After transplantation, different types of host cells migrate to the decellularized tissues. Histological and immunohistochemical assessment of the different bioscaffolds after implantation reveals the migration of parenchymal cells, angiogenesis, as well as the invasion of inflammatory and giant foreign cells. In this review, the events after transplantation including angiogenesis, scaffold degradation, and the presence of immune and tissue-specific progenitor cells in the decellularized scaffolds in various hosts, are discussed.
Collapse
Affiliation(s)
- Tahereh Talaei-Khozani
- Histotomorphometry and stereology research center, Shiraz University of Medical Sciences, Shiraz, Iran; Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Yaghoubi
- Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Fang L, Mei J, Yao B, Liu J, Liu P, Wang X, Zhou J, Lin Z. Hypoxia facilitates proliferation of smooth muscle cells derived from pluripotent stem cells for vascular tissue engineering. J Tissue Eng Regen Med 2022; 16:744-756. [PMID: 35633489 DOI: 10.1002/term.3324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/07/2022]
Abstract
Tissue-engineered blood vessels (TEBVs) show significant therapeutic potential for replacing diseased blood vessels. Vascular smooth muscle cells (VSMCs) derived from human induced pluripotent stem cells (hiPSCs) via embryoid body (EB)-based differentiation, are promising seed cells to construct TEBVs. However, obtaining sufficient high-quality hiPSC-VSMCs remains challenging. Stem cells are located in a niche characterized by hypoxia. Hence, we explored molecular and cellular functions at different induction stages from the EB formation commencement to the end of directed differentiation under normoxic and hypoxic conditions, respectively. Hypoxia enhanced the formation, adhesion and amplification rates of EBs. During directed differentiation, hiPSC-VSMCs exhibited increased cell viability under hypoxic conditions. Moreover, seeding hypoxia-pretreated cells on biodegradable scaffolds, facilitated collagen I and elastin secretion, which has significant application value for TEBV development. Hence, we proposed that hypoxic treatment during differentiation effectively induces proliferative hiPSC-VSMCs, expanding high-quality seed cell sources for TEBV construction.
Collapse
Affiliation(s)
- Lijun Fang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jingyi Mei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Boqian Yao
- Songshan Lake Central Hospital of Dongguan City, Dongguan, Guangdong, China
| | - Jiang Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.,Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guanzhou, China
| | - Peng Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Xichun Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiahui Zhou
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China
| | - Zhanyi Lin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Yaghoubi A, Azarpira N, Karbalay-Doust S, Daneshi S, Vojdani Z, Talaei-Khozani T. Prednisolone and mesenchymal stem cell preloading protect liver cell migration and mitigate extracellular matrix modification in transplanted decellularized rat liver. Stem Cell Res Ther 2022; 13:36. [PMID: 35090559 PMCID: PMC8800282 DOI: 10.1186/s13287-022-02711-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Regenerative medicine provides promising approaches for treating chronic liver diseases. Previous studies indicate that decellularized liver architecture is damaged by invading non-hepatic inflammatory cells. This study aimed to use anti-inflammatory and regenerative potency of bone marrow-derived mesenchymal stem cells (BM-MSC) and prednisolone for reducing fibrosis and balancing inflammatory cell migration into the decellularized liver scaffold. MATERIAL AND METHOD The liver was decellularized by perfusing Sodium Lauryl Ether Sulfate (SLES), and nuclei depletion and extracellular matrix (ECM) retention were confirmed by DNA quantification, histochemical, and immunohistochemical assessments. Scaffolds were loaded with BM-MSCs, prednisolone, or a combination of both, implanted at the anatomical place in the rat partial hepatectomized and followed up for 2 and 4 weeks. RESULTS Labeled-MSCs were traced in the transplanted scaffolds; however, they did not migrate into the intact liver. Immunohistochemistry showed that the hepatoblasts, cholangiocytes, stellate, and oval cells invaded into all the scaffolds. Bile ducts were more abundant in the border of the scaffolds and intact liver. Stereological assessments showed a significant reduction in the number of lymphocytes and neutrophils in prednisolone-loaded scaffolds. The regeneration process and angiogenesis were significantly higher in the group treated with cell/prednisolone-loaded bioscaffolds. Collagen fibers were significantly reduced in the scaffolds pre-treated with cell/prednisolone, prednisolone, or BM-MSCs, compared to the control group. CONCLUSION Loading prednisolone into the scaffolds can be a worthy approach to restrict inflammation after transplantation. Although pre-loading of the scaffolds with a combination of cells/prednisolone could not alleviate inflammation, it played an important role in regeneration and angiogenesis.
Collapse
Affiliation(s)
- Atefeh Yaghoubi
- Tissue Engineering Lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplantation Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saied Karbalay-Doust
- Stereology and Morphometry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy Department, Shiraz medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Daneshi
- Stereology and Morphometry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Vojdani
- Tissue Engineering Lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Tissue Engineering Lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Craig DJ, James AW, Wang Y, Tavian M, Crisan M, Péault BM. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:35-43. [PMID: 35641167 PMCID: PMC8895497 DOI: 10.1093/stcltm/szab001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
The vascular wall is comprised of distinct layers controlling angiogenesis, blood flow, vessel anchorage within organs, and cell and molecule transit between blood and tissues. Moreover, some blood vessels are home to essential stem-like cells, a classic example being the existence in the embryo of hemogenic endothelial cells at the origin of definitive hematopoiesis. In recent years, microvascular pericytes and adventitial perivascular cells were observed to include multi-lineage progenitor cells involved not only in organ turnover and regeneration but also in pathologic remodeling, including fibrosis and atherosclerosis. These perivascular mesodermal elements were identified as native forerunners of mesenchymal stem cells. We have presented in this brief review our current knowledge on vessel wall-associated tissue remodeling cells with respect to discriminating phenotypes, functional diversity in health and disease, and potential therapeutic interest.
Collapse
Affiliation(s)
- David J Craig
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Mihaela Crisan
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Bruno M Péault
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Corresponding author: Bruno Péault, PhD, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, 615 Charles E. Young Drive South, Los Angeles, CA 90095-7358, USA.
| |
Collapse
|
7
|
Translational considerations for adipose-derived biological scaffolds for soft tissue repair. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
8
|
Xiang K, Catanzaro JN, Elayi C, Esquer Garrigos Z, Sohail MR. Antibiotic-Eluting Envelopes to Prevent Cardiac-Implantable Electronic Device Infection: Past, Present, and Future. Cureus 2021; 13:e13088. [PMID: 33728111 PMCID: PMC7948693 DOI: 10.7759/cureus.13088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective: Cardiac-implantable electronic device (CIED) infections are associated with significant morbidity and mortality. In this review, we describe the risk factors and pathogenesis of CIED infections and review the rationale and the evidence for the use of antibiotic-eluting envelopes (ABEs) in patients at increased risk for CIED infections. Findings: The majority of CIED infections are caused by staphylococci that involve generator pocket and occur due to contamination of the device or the pocket tissues at the time of implantation. Clinical trials have shown that extending the duration of post-operative systemic antibacterial therapy is not beneficial in reducing CIED infection rate. However, ABEs that reduce device migration after implantation and provide sustained local delivery of prophylactic antibiotics at the pocket site, may provide benefit in reducing infection. Currently, there are two types of commercially available CIED envelope devices in the United States. The first ABE device (TYRX™, Medtronic Inc., Monmouth Junction, NJ) is composed of a synthetic absorbable mesh envelope that elutes minocycline and rifampin and has been shown to reduce CIED pocket infections in a large multi-center randomized clinical trial. The second ABE device (CanGaroo-G™, Aziyo Biologics, Silver Spring, MD) is composed of decellularized extracellular matrix (ECM) and was originally designed to stabilize the device within the pocket, limiting risk for migration or erosion, and providing a substrate for tissue ingrowth in a preclinical study. This device has shown promising results in a preclinical study with local delivery of gentamicin. Compared with artificial materials, such as synthetic surgical mesh, biologic ECM has been shown to foster greater tissue integration and vascular ingrowth, a reduced inflammatory response, and more rapid clearance of bacteria. Conclusions and Relevance: ABE devices provide sustained local delivery of antibiotics at the generator pocket site and appear beneficial in reducing CIED pocket infections. Given the continued increase in the use of CIED therapy and resultant infectious complications, innovative approaches to infection prevention are critical.
Collapse
Affiliation(s)
- Kun Xiang
- Cardiology, University of Florida College of Medicine, Gainesville, USA
| | - John N Catanzaro
- Cardiology, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Claude Elayi
- Cardiology, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Zerelda Esquer Garrigos
- Internal Medicine/Infectious Disease, Mayo Clinic College of Medicine, University of Mississippi Medical Center, Rochester, USA
| | - Muhammad R Sohail
- Cardiovascular Infectious Diseases, Baylor College of Medicine, Houston, USA
| |
Collapse
|
9
|
Wu B, Jin L, Ding K, Zhou Y, Yang L, Lei Y, Guo Y, Wang Y. Extracellular matrix coating improves the biocompatibility of polymeric heart valves. J Mater Chem B 2020; 8:10616-10629. [PMID: 33146226 DOI: 10.1039/d0tb01884h] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Prosthetic heart valve replacement is an effective therapy for patients with valvular heart disease. New-type polymer materials provide potential choices of material for preparing prosthetic heart valves. In this study, we focused on enhancing the biocompatibility of polystyrene-block-isobutylene-block-styrene (SIBS) by surface modification with an extracellular matrix (ECM). Experimental results demonstrated that the ECM coating increased the adsorption resistance against protein and platelets. SIBS coated with an ECM adsorbed much less bovine serum albumin and fibrinogen (5.38 μg cm-2 and 31.53 μg cm-2, respectively) than the original material (90.84 μg cm-2 and 132.38 μg cm-2, respectively). The relative platelet adsorption of the ECM-modified SIBS was lower than that of SIBS (0.04 versus 0.10). Moreover, the surface coating could also reduce endothelial cytotoxicity, suppress the immune response, and potentially induce tissue regeneration. In conclusion, ECM coating improved the biocompatibility of SIBS effectively.
Collapse
Affiliation(s)
- Binggang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China. and Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, P. R. China
| | - Linhe Jin
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Kailei Ding
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Yonghua Zhou
- Beijing Huiyu Biomedical Technologies LLC, 1707 street, Chaoyang District, Beijing 100000, P. R. China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Yang Lei
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| |
Collapse
|
10
|
Pereira I, Pereira JE, Maltez L, Rodrigues A, Rodrigues C, Oliveira M, Silva DM, Caseiro AR, Prada J, Maurício AC, Santos JD, Gama M. Regeneration of critical-sized defects, in a goat model, using a dextrin-based hydrogel associated with granular synthetic bone substitute. Regen Biomater 2020; 8:rbaa036. [PMID: 33732486 PMCID: PMC7947577 DOI: 10.1093/rb/rbaa036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
The development of injectable bone substitutes (IBS) have obtained great importance in the bone regeneration field, as a strategy to reach hardly accessible defects using minimally invasive techniques and able to fit to irregular topographies. In this scenario, the association of injectable hydrogels and bone graft granules is emerging as a well-established trend. Particularly, in situ forming hydrogels have arisen as a new IBS generation. An in situ forming and injectable dextrin-based hydrogel (HG) was developed, aiming to act as a carrier of granular bone substitutes and bioactive agents. In this work, the HG was associated to a granular bone substitute (Bonelike®) and implanted in goat critical-sized calvarial defects (14 mm) for 3, 6 and 12 weeks. The results showed that HG improved the handling properties of the Bonelike® granules and did not affect its osteoconductive features, neither impairing the bone regeneration process. Human multipotent mesenchymal stromal cells from the umbilical cord, extracellular matrix hydrolysates and the pro-angiogenic peptide LLKKK18 were also combined with the IBS. These bioactive agents did not enhance the new bone formation significantly under the conditions tested, according to micro-computed tomography and histological analysis.
Collapse
Affiliation(s)
- Isabel Pereira
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
- Correspondence address. CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal. Tel: +351-253-604-418; E-mail:
| | - José Eduardo Pereira
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Luís Maltez
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Alexandra Rodrigues
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Catarina Rodrigues
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Manuela Oliveira
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Dina M Silva
- Biosckin, Molecular and Cell Therapies S.A., Laboratório Criovida, TecMaia, Rua Engenheiro Frederico Ulrich 2650, Moreira da Maia 4470-605, Portugal
| | - Ana Rita Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto 4051-401 Portugal
- Centro de Investigação Vasco da Gama (CIVG)/Escola Universitária Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, n.° 197 Lordemão, Coimbra 3020-210, Portugal
| | - Justina Prada
- CECAV, Animal and Veterinary Research Centre, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real 5001-801, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto 4051-401 Portugal
| | - José Domingos Santos
- REQUIMTE/LAQV, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr Roberto Frias, Porto 4200-495, Portugal
| | - Miguel Gama
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| |
Collapse
|
11
|
Vessel Wall-Derived Mesenchymal Stromal Cells Share Similar Differentiation Potential and Immunomodulatory Properties with Bone Marrow-Derived Stromal Cells. Stem Cells Int 2020; 2020:8847038. [PMID: 33144864 PMCID: PMC7596426 DOI: 10.1155/2020/8847038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/04/2020] [Accepted: 10/09/2020] [Indexed: 01/07/2023] Open
Abstract
Purpose This study is aimed at investigating the phenotype, differentiation potential, immunomodulatory properties, and responsiveness of saphenous vein vessel wall-derived mesenchymal stromal cells (SV-MSCs) to various TLR ligands and proinflammatory cytokines, as well as comparing their features to those of their bone marrow-derived counterparts (BM-MSCs). Methods SV-MSCs were isolated by enzymatic digestion of the saphenous vein vessel wall. Phenotype analysis was carried out by flow cytometry and microscopy, whereas adipogenic, chondrogenic, and osteogenic differentiation potentials were tested in in vitro assays. For comparative analysis, the expression of different stemness, proliferation, and differentiation-related genes was determined by Affymetrix gene array. To compare the immunomodulatory properties of SV-MSCs and BM-MSCs, mixed lymphocyte reaction was applied. To investigate their responses to various activating stimuli, MSCs were treated with TLR ligands (LPS, PolyI:C) or proinflammatory cytokines (TNFα, IL-1β, IFNγ), and the expression of various early innate immune response-related genes was assessed by qPCR, while secretion of selected cytokines and chemokines was measured by ELISA. Results The isolated SV-MSCs were able to differentiate into bone, fat, and cartilage cells/direction in vitro. SV-MSCs expressed the most important MSC markers (CD29, CD44, CD73, CD90, and CD105) and shared almost identical phenotypic characteristics with BM-MSCs. Their gene expression pattern and activation pathways were close to those of BM-MSCs. SV-MSCs showed better immunosuppressive activity inhibiting phytohemagglutinin-induced T lymphocyte proliferation in vitro than BM-MSCs. Cellular responses to treatments mimicking inflammatory conditions were comparable in the bone marrow- and saphenous vein-derived MSCs. Namely, similar to BM-MSCs, SV-MSCs secreted increased amount of IL-6 and IL-8 after 12- or 24-hour treatment with LPS, PolyI:C, TNFα, or IL-1β, compared to untreated controls. Interestingly, a different CXCL-10/IP-10 secretion pattern could be observed under inflammatory conditions in the two types of MSCs. Conclusion Based on our results, cells isolated from saphenous vein vessel wall fulfilled the ISCT's (International Society for Cellular Therapy) criteria for multipotent mesenchymal stromal cells, and no significant differences in the phenotype, gene expression pattern, and responsiveness to inflammatory stimuli could be observed between BM-MSCs and SV-MSCs, while the latter cells have more potent immunosuppressive activity in vitro. Further functional assays have to be performed to reveal whether SV-MSCs could be useful for certain regenerative therapeutic applications or tissue engineering purposes.
Collapse
|
12
|
A novel chemotactic factor derived from the extracellular matrix protein decorin recruits mesenchymal stromal cells in vitro and in vivo. PLoS One 2020; 15:e0235784. [PMID: 32658899 PMCID: PMC7357784 DOI: 10.1371/journal.pone.0235784] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
Soft tissue is composed of cells surrounded by an extracellular matrix that is made up of a diverse array of intricately organized proteins. These distinct components work in concert to maintain homeostasis and respond to tissue damage. During tissue repair, extracellular matrix proteins and their degradation products are known to influence physiological processes such as angiogenesis and inflammation. In this study we developed a discovery platform using a decellularized extracellular matrix biomaterial to identify new chemotrophic factors derived from the extracellular matrix. An in vitro culture of RAW.264 macrophage cells with the biomaterial ovine forestomach matrix led to the identification of a novel ~12 kDa chemotactic factor, termed ‘MayDay’, derived from the N-terminal 31–188 sequence of decorin. The recombinant MayDay protein was shown to be a chemotactic agent for mesenchymal stromal cells in vitro and in vivo. We hypothesize that the macrophage-induced cleavage of decorin, via MMP-12, leads to the release of the chemotactic molecule MayDay, that in turn recruits cells to the site of damaged tissue.
Collapse
|
13
|
Mayo JN, Kauer SD, Brumley MR, Bearden SE. Pericytes improve locomotor recovery after spinal cord injury in male and female neonatal rats. Microcirculation 2020; 27:e12646. [PMID: 32608116 DOI: 10.1111/micc.12646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/06/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE It is not known how activation of the hypoxia-inducible factor (HIF) pathway in pericytes, cells of the microvascular wall, influences new capillary growth. We tested the hypothesis that HIF-activated pericytes promote angiogenesis in a neonatal model of spinal cord injury (SCI). METHODS Human placental pericytes stimulated with cobalt chloride and naïve pericytes were injected into the site of a thoracic hemi-section of the spinal cord in rat pups on postnatal day three (P3). Hindlimb motor recovery and Doppler blood flow perfusion at the site of transection were measured on P10. Immunohistochemistry was used to visualize vessel and neurofilament density for quantification. RESULTS Injection of HIF-activated pericytes resulted in greater vascular density in males but did not result in improved motor function for males or females. Injection of non-HIF-activated pericytes resulted improved motor function recovery in both sexes (males, 2.722 ± 0.31-fold score improvement; females, 3.824 ± 0.58-fold score improvement, P < .05) but produced no significant changes in vessel density. CONCLUSIONS HIF-activated pericytes promote vascular density in males post-SCI. Acute delivery of non-HIF-activated pericytes at the site of injury can improve motor recovery post-SCI.
Collapse
Affiliation(s)
- Jamie N Mayo
- IDEAS 2.0 Centre of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, USA.,Division of Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.,Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Sierra D Kauer
- Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Michele R Brumley
- Department of Psychology, Idaho State University, Pocatello, ID, USA
| | - Shawn E Bearden
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
14
|
Identification of High-Quality Fat Based on Precision Centrifugation in Lipoaspirates Using Marker Floats. Plast Reconstr Surg 2020; 146:541-550. [DOI: 10.1097/prs.0000000000007063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
15
|
Liao J, Xu B, Zhang R, Fan Y, Xie H, Li X. Applications of decellularized materials in tissue engineering: advantages, drawbacks and current improvements, and future perspectives. J Mater Chem B 2020; 8:10023-10049. [PMID: 33053004 DOI: 10.1039/d0tb01534b] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Decellularized materials (DMs) are attracting more and more attention in tissue engineering because of their many unique advantages, and they could be further improved in some aspects through various means.
Collapse
Affiliation(s)
- Jie Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Bo Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Ruihong Zhang
- Department of Research and Teaching
- the Fourth Central Hospital of Baoding City
- Baoding 072350
- China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University and Collaborative Innovation Center of Biotherapy
- Chengdu 610041
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| |
Collapse
|
16
|
Nguyen L, Lu P, Boehm D, Bourke P, Gilmore BF, Hickok NJ, Freeman TA. Cold atmospheric plasma is a viable solution for treating orthopedic infection: a review. Biol Chem 2019; 400:77-86. [PMID: 30138104 DOI: 10.1515/hsz-2018-0235] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022]
Abstract
Bacterial infection and antibiotic resistance are major threats to human health and very few solutions are available to combat this eventuality. A growing number of studies indicate that cold (non-thermal) plasma treatment can be used to prevent or eliminate infection from bacteria, bacterial biofilms, fungi and viruses. Mechanistically, a cold plasma discharge is composed of high-energy electrons that generate short-lived reactive oxygen and nitrogen species which further react to form more stable compounds (NO2, H2O2, NH2Cl and others) depending on the gas mixture and plasma parameters. Cold plasma devices are being developed for medical applications including infection, cancer, plastic surgery applications and more. Thus, in this review we explore the potential utility of cold plasma as a non-antibiotic approach for treating post-surgical orthopedic infections.
Collapse
Affiliation(s)
- Ly Nguyen
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107-5099, USA
| | - Peng Lu
- College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Daniela Boehm
- College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Paula Bourke
- College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Brendan F Gilmore
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107-5099, USA
| | - Theresa A Freeman
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107-5099, USA
| |
Collapse
|
17
|
Marcinczyk M, Dunn A, Haas G, Madsen J, Scheidt R, Patel K, Talovic M, Garg K. The Effect of Laminin-111 Hydrogels on Muscle Regeneration in a Murine Model of Injury. Tissue Eng Part A 2019; 25:1001-1012. [PMID: 30426851 PMCID: PMC9839345 DOI: 10.1089/ten.tea.2018.0200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
IMPACT STATEMENT Extremity injuries make up the most common survivable injuries in vehicular accidents and modern military conflicts. A majority of these injuries involve volumetric muscle loss (VML). The potential for donor site morbidity may limit the clinical use of autologous muscle grafts for VML injuries. Treatments that can improve the regeneration of functional muscle tissue are critically needed to improve limb salvage and reduce the rate of delayed amputations. The development of a laminin-111-enriched fibrin hydrogel will offer a potentially transformative and "off-the-shelf" clinically relevant therapy for functional skeletal muscle regeneration.
Collapse
Affiliation(s)
- Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Josh Madsen
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Robert Scheidt
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri.,Address correspondence to: Koyal Garg, PhD, Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, 3507 Lindell Boulevard, St. Louis, MO 63103
| |
Collapse
|
18
|
Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019; 37:1246-1262. [PMID: 30604468 DOI: 10.1002/jor.24212] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 02/04/2023]
Abstract
Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration. These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes, fibroadipogenic progenitors, side population cells, and CD133+ stem cells. However, several challenges associated with their isolation, availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal muscle injury and disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246-1262, 2019.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
19
|
|
20
|
Andreeva ER, Udartseva OO, Zhidkova OV, Buravkov SV, Ezdakova MI, Buravkova LB. IFN‐gamma priming of adipose‐derived stromal cells at “physiological” hypoxia. J Cell Physiol 2017; 233:1535-1547. [DOI: 10.1002/jcp.26046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Elena R. Andreeva
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Olga O. Udartseva
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Olga V. Zhidkova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | | | - Maria I. Ezdakova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Ludmila B. Buravkova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| |
Collapse
|
21
|
Vashaghian M, Zaat SJ, Smit TH, Roovers JP. Biomimetic implants for pelvic floor repair. Neurourol Urodyn 2017; 37:566-580. [PMID: 28799675 DOI: 10.1002/nau.23367] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/15/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Polypropylene implants are used for the reconstructive surgery of urogynaecological disorders like pelvic organ prolapse, but severe complications associated with their use have been reported. There is evidence that surface properties and a difference in mechanical stiffness between the implant and the host tissue contribute to these adverse events. Electrospinning is an innovative engineering alternative that provides a biomimetic microstructure for implants, resulting in a different mechano-biological performance. AIM The main objective of this review is to inform about the potential of electrospun matrices as an alternative modality for pelvic floor repair. METHODS Publications with the following studies of electrospun matrices were reviewed: (i) the technique; (ii) in vitro use for soft tissue engineering; (iii) in vivo use for reconstruction of soft tissues in animals; and (iv) clinical use in humans. RESULTS Electrospun matrices provide a synthetic mimic of natural extracellular matrix (ECM), favoring cellular attachment, proliferation and matrix deposition, through which a proper, low-inflammatory tissue-implant interaction can be established. Electrospun sheets can also be created with sufficient mechanical strength and stiffness for usage in prolapse surgery. CONCLUSION Electrospun matrices mimic the structural topography of the extracellular matrix and can be functionalized for better biological performance. As such, they have great potential for the next generation of urogynecological implants. However, their long-term safety and efficacy must still be established in vivo.
Collapse
Affiliation(s)
- Mahshid Vashaghian
- Department of Obstetrics & Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sebastianus J Zaat
- Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Theodoor H Smit
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan-Paul Roovers
- Department of Obstetrics & Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Gaffney L, Wrona EA, Freytes DO. Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds. ACS Biomater Sci Eng 2017. [DOI: 10.1021/acsbiomaterials.7b00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Lewis Gaffney
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Emily A. Wrona
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
23
|
Angelozzi M, Penolazzi L, Mazzitelli S, Lambertini E, Lolli A, Piva R, Nastruzzi C. Dedifferentiated Chondrocytes in Composite Microfibers As Tool for Cartilage Repair. Front Bioeng Biotechnol 2017; 5:35. [PMID: 28660185 PMCID: PMC5468460 DOI: 10.3389/fbioe.2017.00035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022] Open
Abstract
Tissue engineering (TE) approaches using biomaterials have gain important roles in the regeneration of cartilage. This paper describes the production by microfluidics of alginate-based microfibers containing both extracellular matrix (ECM)-derived biomaterials and chondrocytes. As ECM components gelatin or decellularized urinary bladder matrix (UBM) were investigated. The effectiveness of the composite microfibers has been tested to modulate the behavior and redifferentiation of dedifferentiated chondrocytes. The complete redifferentiation, at the single-cell level, of the chondrocytes, without cell aggregate formation, was observed after 14 days of cell culture. Specific chondrogenic markers and high cellular secretory activity was observed in embedded cells. Notably, no sign of collagen type 10 deposition was determined. The obtained data suggest that dedifferentiated chondrocytes regain a functional chondrocyte phenotype when embedded in appropriate 3D scaffold based on alginate plus gelatin or UBM. The proposed scaffolds are indeed valuable to form a cellular microenvironment mimicking the in vivo ECM, opening the way to their use in cartilage TE.
Collapse
Affiliation(s)
- Marco Angelozzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Stefania Mazzitelli
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Claudio Nastruzzi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
24
|
Morris AH, Stamer DK, Kyriakides TR. The host response to naturally-derived extracellular matrix biomaterials. Semin Immunol 2017; 29:72-91. [PMID: 28274693 DOI: 10.1016/j.smim.2017.01.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 12/13/2022]
Abstract
Biomaterials based on natural materials including decellularized tissues and tissue-derived hydrogels are becoming more widely used for clinical applications. Because of their native composition and structure, these biomaterials induce a distinct form of the foreign body response that differs from that of non-native biomaterials. Differences include direct interactions with cells via preserved moieties as well as the ability to undergo remodeling. Moreover, these biomaterials could elicit adaptive immune responses due to the presence of modified native molecules. Therefore, these biomaterials present unique challenges in terms of understanding the progression of the foreign body response. This review covers this response to natural materials including natural polymers, decellularized tissues, cell-derived matrix, tissue derived hydrogels, and biohybrid materials. With the expansion of the fields of regenerative medicine and tissue engineering, the current repertoire of biomaterials has also expanded and requires continuous investigation of the responses they elicit.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States
| | - D K Stamer
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - T R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Department of Pathology, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.
| |
Collapse
|
25
|
Tukmachev D, Forostyak S, Koci Z, Zaviskova K, Vackova I, Vyborny K, Sandvig I, Sandvig A, Medberry CJ, Badylak SF, Sykova E, Kubinova S. Injectable Extracellular Matrix Hydrogels as Scaffolds for Spinal Cord Injury Repair. Tissue Eng Part A 2016; 22:306-17. [PMID: 26729284 DOI: 10.1089/ten.tea.2015.0422] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Restoration of lost neuronal function after spinal cord injury (SCI) still remains a big challenge for current medicine. One important repair strategy is bridging the SCI lesion with a supportive and stimulatory milieu that would enable axonal rewiring. Injectable extracellular matrix (ECM)-derived hydrogels have been recently reported to have neurotrophic potential in vitro. In this study, we evaluated the presumed neuroregenerative properties of ECM hydrogels in vivo in the acute model of SCI. ECM hydrogels were prepared by decellularization of porcine spinal cord (SC) or porcine urinary bladder (UB), and injected into a spinal cord hemisection cavity. Histological analysis and real-time qPCR were performed at 2, 4, and 8 weeks postinjection. Both types of hydrogels integrated into the lesion and stimulated neovascularization and axonal ingrowth into the lesion. On the other hand, massive infiltration of macrophages into the lesion and rapid hydrogel degradation did not prevent cyst formation, which progressively developed over 8 weeks. No significant differences were found between SC-ECM and UB-ECM. Gene expression analysis revealed significant downregulation of genes related to immune response and inflammation in both hydrogel types at 2 weeks post SCI. A combination of human mesenchymal stem cells with SC-ECM did not further promote ingrowth of axons and blood vessels into the lesion, when compared with the SC-ECM hydrogel alone. In conclusion, both ECM hydrogels bridged the lesion cavity, modulated the innate immune response, and provided the benefit of a stimulatory substrate for in vivo neural tissue regeneration. However, fast hydrogel degradation might be a limiting factor for the use of native ECM hydrogels in the treatment of acute SCI.
Collapse
Affiliation(s)
- Dmitry Tukmachev
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Serhiy Forostyak
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Zuzana Koci
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Kristyna Zaviskova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Irena Vackova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic
| | - Karel Vyborny
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Ioanna Sandvig
- 3 Department of Neuroscience, Norwegian University of Science and Technology , Trondheim, Norway .,4 John Van Geest Centre for Brain Repair, School of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom
| | - Axel Sandvig
- 3 Department of Neuroscience, Norwegian University of Science and Technology , Trondheim, Norway .,5 Division of Pharmacology and Clinical Neuroscience, Department of Neurosurgery, Umeå University , Umeå, Sweden
| | | | - Stephen F Badylak
- 6 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania
| | - Eva Sykova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Sarka Kubinova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic
| |
Collapse
|
26
|
Badylak SF, Dziki JL, Sicari BM, Ambrosio F, Boninger ML. Mechanisms by which acellular biologic scaffolds promote functional skeletal muscle restoration. Biomaterials 2016; 103:128-136. [DOI: 10.1016/j.biomaterials.2016.06.047] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
|
27
|
Burnsed OA, Schwartz Z, Marchand KO, Hyzy SL, Olivares-Navarrete R, Boyan BD. Hydrogels derived from cartilage matrices promote induction of human mesenchymal stem cell chondrogenic differentiation. Acta Biomater 2016; 43:139-149. [PMID: 27449339 DOI: 10.1016/j.actbio.2016.07.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 07/17/2016] [Accepted: 07/19/2016] [Indexed: 12/01/2022]
Abstract
UNLABELLED Limited supplies of healthy autologous or allogeneic cartilage sources have inspired a growing interest in xenogeneic cartilage matrices as biological scaffolds for cartilage tissue engineering. The objectives of this study were to determine if shark and pig cartilage extracellular matrix (ECM) hydrogels can stimulate chondrocytic differentiation of mesenchymal stem cells (MSCs) without exogenous growth factors and to determine if the soluble factors retained by these ECM hydrogels are responsible. Human MSCs cultured on hydrogels from shark skull cartilage, pig articular cartilage, and pig auricular cartilage ECM had increased expression of chondrocyte markers and decreased secretion of angiogenic factors VEGF-A and FGF2 in comparison to MSCs cultured on tissue culture polystyrene (TCPS) at one week. MSCs grown on shark ECM gels had decreased type-1 collagen mRNA as compared to all other groups. Degradation products of the cartilage ECM gels and soluble factors released by the matrices increased chondrogenic and decreased angiogenic mRNA levels, indicating that the processed ECM retains biochemically active proteins that can stimulate chondrogenic differentiation. In conclusion, this work supports the use of cartilage matrix-derived hydrogels for chondrogenic differentiation of MSCs and cartilage tissue engineering. Longer-term studies and positive controls will be needed to support these results to definitively demonstrate stimulation of chondrocyte differentiation, and particularly to verify that calcification without endochondral ossification does not occur as it does in shark cartilage. STATEMENT OF SIGNIFICANCE The objectives of this study were to determine if shark and pig cartilage extracellular matrix (ECM) hydrogels can stimulate chondrocytic differentiation of mesenchymal stem cells (MSCs) without exogenous growth factors and to determine if the soluble factors retained by these ECM hydrogels are responsible for this induction. Sharks are an especially interesting model for cartilage regeneration because their entire skeleton is composed of cartilage and they do not undergo endochondral ossification. Culturing human MSCs on porcine and shark cartilage ECM gels directly, with ECM gel conditioned media, or degradation products increased mRNA levels of chondrogenic factors while decreasing angiogenic factors. These studies indicate that xenogeneic cartilage ECMs have potential as biodegradable scaffolds capable of stimulating chondrogenesis while preventing angiogenesis for regenerative medicine applications and that ECM species selection can yield differential effects.
Collapse
Affiliation(s)
- Olivia A Burnsed
- Wallace H. Coulter Department of Biomedical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Katherine O Marchand
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sharon L Hyzy
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Barbara D Boyan
- Wallace H. Coulter Department of Biomedical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
28
|
Eisenhauer P, Chernets N, Song Y, Dobrynin D, Pleshko N, Steinbeck MJ, Freeman TA. Chemical modification of extracellular matrix by cold atmospheric plasma-generated reactive species affects chondrogenesis and bone formation. J Tissue Eng Regen Med 2016; 10:772-82. [PMID: 27510797 DOI: 10.1002/term.2224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/15/2016] [Accepted: 04/19/2016] [Indexed: 12/17/2022]
Abstract
The goal of this study was to investigate whether cold plasma generated by dielectric barrier discharge (DBD) modifies extracellular matrices (ECM) to influence chondrogenesis and endochondral ossification. Replacement of cartilage by bone during endochondral ossification is essential in fetal skeletal development, bone growth and fracture healing. Regulation of this process by the ECM occurs through matrix remodelling, involving a variety of cell attachment molecules and growth factors, which influence cell morphology and protein expression. The commercially available ECM, Matrigel, was treated with microsecond or nanosecond pulsed (μsp or nsp, respectively) DBD frequencies conditions at the equivalent frequencies (1 kHz) or power (~1 W). Recombinant human bone morphogenetic protein-2 was added and the mixture subcutaneously injected into mice to simulate ectopic endochondral ossification. Two weeks later, the masses were extracted and analysed by microcomputed tomography. A significant increase in bone formation was observed in Matrigel treated with μsp DBD compared with control, while a significant decrease in bone formation was observed for both nsp treatments. Histological and immunohistochemical analysis showed Matrigel treated with μsp plasma increased the number of invading cells, the amount of vascular endothelial growth factor and chondrogenesis while the opposite was true for Matrigel treated with nsp plasma. In support of the in vivo Matrigel study, 10 T1/2 cells cultured in vitro on μsp DBD-treated type I collagen showed increased expression of adhesion proteins and activation of survival pathways, which decreased with nsp plasma treatments. These results indicate DBD modification of ECM can influence cellular behaviours to accelerate or inhibit chondrogenesis and endochondral ossification. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Peter Eisenhauer
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Natalie Chernets
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - You Song
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Danil Dobrynin
- Drexel Plasma Institute, Drexel University, Philadelphia, PA, USA
| | - Nancy Pleshko
- Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Marla J Steinbeck
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Theresa A Freeman
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA. .,Drexel Plasma Institute, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Keane TJ, DeWard A, Londono R, Saldin LT, Castleton AA, Carey L, Nieponice A, Lagasse E, Badylak SF. Tissue-Specific Effects of Esophageal Extracellular Matrix. Tissue Eng Part A 2016; 21:2293-300. [PMID: 26192009 DOI: 10.1089/ten.tea.2015.0322] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Biologic scaffolds composed of extracellular matrix (ECM) have been used to facilitate repair or remodeling of numerous tissues, including the esophagus. The theoretically ideal scaffold for tissue repair is the ECM derived from the particular tissue to be treated, that is, site-specific or homologous ECM. The preference or potential advantage for the use of site-specific ECM remains unknown in the esophageal location. The objective of the present study was to characterize the in vitro cellular response and in vivo host response to a homologous esophageal ECM (eECM) versus nonhomologous ECMs derived from small intestinal submucosa and urinary bladder. The in vitro response of esophageal stem cells was characterized by migration, proliferation, and three-dimensional (3D) organoid formation assays. The in vivo remodeling response was evaluated in a rat model of esophageal mucosal resection. Results of the study showed that the eECM retains favorable tissue-specific characteristics that enhance the migration of esophageal stem cells and supports the formation of 3D organoids to a greater extent than heterologous ECMs. Implantation of eECM facilitates the remodeling of esophageal mucosa following mucosal resection, but no distinct advantage versus heterologous ECM could be identified.
Collapse
Affiliation(s)
- Timothy J Keane
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Aaron DeWard
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,3 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Ricardo Londono
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,4 School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lindsey T Saldin
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Arthur A Castleton
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania
| | - Lisa Carey
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Alejandro Nieponice
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,5 Hospital Universitario , Fundación Favaloro, Buenos Aires, Argentina
| | - Eric Lagasse
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,3 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Stephen F Badylak
- 1 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania.,6 Department of Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
30
|
Li CS, Zhang X, Péault B, Jiang J, Ting K, Soo C, Zhou YH. Accelerated Chondrogenic Differentiation of Human Perivascular Stem Cells with NELL-1. Tissue Eng Part A 2016; 22:272-85. [PMID: 26700847 PMCID: PMC4779324 DOI: 10.1089/ten.tea.2015.0250] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/08/2015] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis is the leading cause of disability in the US. Consequently, there is a pressing need for restoring the structural and functional properties of diseased articular cartilage. Yet the search for the right combination of proper target cells and growth factors for cartilage regeneration remains challenging. In this study, we first tested the intrinsic chondrogenic differentiation ability of human perivascular stem cells (hPSCs), a novel source of mesenchymal stem cells (MSCs) isolated by fluorescence-activated cell sorting (FACS) from human adipose tissue. A putative prochondrogenic growth factor, NEL-like molecule-1 (NELL-1), was added to the hPSC pellets to upregulate gene expression of chondrogenic markers, including AGGRECAN, COLLAGEN II, and COMP. Furthermore, the addition of NELL-1 to a transforming growth factor beta 3 (TGF-β3) + bone morphogenetic protein-6 (BMP-6) "cocktail" resulted in the best combinatorial stimulation in accelerating the chondrogenic differentiation of hPSCs, as evidenced by increased gene and protein expression of chondrogenic markers in a shortened induction time without elevating expression of hypertrophic, fibrotic, and osteogenic markers. Mechanistically, this acceleration rendered by NELL-1 may be partially attributed to NELL-1's upregulation of BMP receptors and TGF-β receptor type I in hPSCs for increased responsiveness to BMPs + TGF-βs. In conclusion, lipoaspirate-derived hPSCs present a novel and abundant cell source of MSCs for cartilage regeneration, and the combinatorial application of NELL-1, TGF-β3, and BMP-6 with hPSCs may remarkably enhance and accelerate cartilage repair.
Collapse
Affiliation(s)
- Chen-Shuang Li
- Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, P.R. China
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Bruno Péault
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
- Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jie Jiang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, P.R. China
| |
Collapse
|
31
|
Poghosyan T, Catry J, Luong-Nguyen M, Bruneval P, Domet T, Arakelian L, Sfeir R, Michaud L, Vanneaux V, Gottrand F, Larghero J, Cattan P. Esophageal tissue engineering: Current status and perspectives. J Visc Surg 2015; 153:21-9. [PMID: 26711880 DOI: 10.1016/j.jviscsurg.2015.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Tissue engineering, which consists of the combination and in vivo implantation of elements required for tissue remodeling toward a specific organ phenotype, could be an alternative for classical techniques of esophageal replacement. The current hybrid approach entails creation of an esophageal substitute composed of an acellular matrix and autologous epithelial and muscle cells provides the most successful results. Current research is based on the use of mesenchymal stem cells, whose potential for differentiation and proangioogenic, immune-modulator and anti-inflammatory properties are important assets. In the near future, esophageal substitutes could be constructed from acellular "intelligent matrices" that contain the molecules necessary for tissue regeneration; this should allow circumvention of the implantation step and still obtain standardized in vivo biological responses. At present, tissue engineering applications to esophageal replacement are limited to enlargement plasties with absorbable, non-cellular matrices. Nevertheless, the application of existing clinical techniques for replacement of other organs by tissue engineering in combination with a multiplication of translational research protocols for esophageal replacement in large animals should soon pave the way for health agencies to authorize clinical trials.
Collapse
Affiliation(s)
- T Poghosyan
- Service de chirurgie digestive, oncologique et métabolique, hôpital Ambroise-Paré, AP-HP, 92100 Boulogne-Billancourt, France; CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - J Catry
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - M Luong-Nguyen
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - P Bruneval
- Service d'anatomopathologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - T Domet
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - L Arakelian
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - R Sfeir
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - L Michaud
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - V Vanneaux
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - F Gottrand
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - J Larghero
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - P Cattan
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France.
| |
Collapse
|
32
|
Sicari BM, Dziki JL, Badylak SF. Strategies for functional bioscaffold-based skeletal muscle reconstruction. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:256. [PMID: 26605302 DOI: 10.3978/j.issn.2305-5839.2015.09.45] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue engineering and regenerative medicine-based strategies for the reconstruction of functional skeletal muscle tissue have included cellular and acellular approaches. The use of acellular biologic scaffold material as a treatment for volumetric muscle loss (VML) in five patients has recently been reported with a generally favorable outcome. Further studies are necessary for a better understanding of the mechanism(s) behind acellular bioscaffold-mediated skeletal muscle repair, and for combination cell-based/bioscaffold based approaches. The present overview highlights the current thinking on bioscaffold-based remodeling including the associated mechanisms and the future of scaffold-based skeletal muscle reconstruction.
Collapse
Affiliation(s)
- Brian M Sicari
- 1 McGowan Institute for Regenerative Medicine, 2 Department of Surgery, 3 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna L Dziki
- 1 McGowan Institute for Regenerative Medicine, 2 Department of Surgery, 3 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen F Badylak
- 1 McGowan Institute for Regenerative Medicine, 2 Department of Surgery, 3 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Sicari BM, Londono R, Badylak SF. Strategies for skeletal muscle tissue engineering: seed vs. soil. J Mater Chem B 2015; 3:7881-7895. [PMID: 32262901 DOI: 10.1039/c5tb01714a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The most commonly used tissue engineering approach includes the ex vivo combination of site-appropriate cell(s) and scaffold material(s) to create three-dimensional constructs for tissue replacement or reconstruction. These three-dimensional combinations are typically subjected to a period of culture and conditioning (i.e., self-assembly and maturation) to promote the development of ex vivo constructs which closely mimic native target tissue. This cell-based approach is challenged by the host response to the engineered tissue construct following surgical implantation. As an alternative to the cell-based approach, acellular biologic scaffolds attract endogenous cells and remodel into partially functional mimics of native tissue upon implantation. The present review examines cell-types (i.e., seed), scaffold materials (i.e., soil), and challenges associated with functional tissue engineering. Skeletal muscle is used as the target tissue prototype but the discussed principles will largely apply to most body systems.
Collapse
Affiliation(s)
- Brian M Sicari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Suite 300, 450 Technology Drive, Pittsburgh, PA 15218, USA.
| | | | | |
Collapse
|
34
|
Andreeva ER, Pogodina MV, Buravkova LB. Hypoxic stress as an activation trigger of multipotent mesenchymal stromal cells. ACTA ACUST UNITED AC 2015. [DOI: 10.1134/s0362119715020024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Wu J, Ding Q, Dutta A, Wang Y, Huang YH, Weng H, Tang L, Hong Y. An injectable extracellular matrix derived hydrogel for meniscus repair and regeneration. Acta Biomater 2015; 16:49-59. [PMID: 25644450 DOI: 10.1016/j.actbio.2015.01.027] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/23/2014] [Accepted: 01/20/2015] [Indexed: 02/07/2023]
Abstract
Tissue-derived extracellular matrix (ECM) biomaterials to regenerate the meniscus have gained increasing attention in treating meniscus injuries and diseases, particularly for aged persons and athletes. However, ECM scaffold has poor cell infiltration and can only be implanted using surgical procedures. To overcome these limitations, we developed an injectable ECM hydrogel material from porcine meniscus via modified decellularization and enzymatic digestion. This meniscus-derived ECM hydrogel exhibited a fibrous morphology with tunable compression and initial modulus. It had a good injectability evidenced by syringe injection into mouse subcutaneous tissue. The hydrogel showed good cellular compatibility by promoting the growth of both bovine chondrocytes and mouse 3T3 fibroblasts encapsulated in the hydrogel for 2 weeks. It also promoted cell infiltration as shown in both in vitro cell culture and in vivo mouse subcutaneous implantation. The in vivo study revealed that the ECM hydrogel possessed good tissue compatibility after 7 days of implantation. The results support the great potential of the newly produced injectable meniscus-derived ECM hydrogel specifically for meniscus repair and regeneration.
Collapse
|
36
|
Faulk DM, Wildemann JD, Badylak SF. Decellularization and cell seeding of whole liver biologic scaffolds composed of extracellular matrix. J Clin Exp Hepatol 2015; 5:69-80. [PMID: 25941434 PMCID: PMC4415199 DOI: 10.1016/j.jceh.2014.03.043] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/03/2014] [Indexed: 12/12/2022] Open
Abstract
The definitive treatment for patients with end-stage liver disease is orthotropic transplantation. However, this option is limited by the disparity between the number of patients needing transplantation and the number of available livers. This issue is becoming more severe as the population ages and as the number of new cases of end-stage liver failure increases. Patients fortunate enough to receive a transplant are required to receive immunosuppressive therapy and must live with the associated morbidity. Whole organ engineering of the liver may offer a solution to this liver donor shortfall. It has been shown that perfusion decellularization of a whole allogeneic or xenogeneic liver generates a three-dimensional ECM scaffold with intact macro and micro architecture of the native liver. A decellularized liver provides an ideal transplantable scaffold with all the necessary ultrastructure and signaling cues for cell attachment, differentiation, vascularization, and function. In this review, an overview of complementary strategies for creating functional liver grafts suitable for transplantation is provided. Early milestones have been met by combining stem and progenitor cells with increasingly complex scaffold materials and culture conditions.
Collapse
Key Words
- BAL, biohybrid artificial liver
- BMC, basement membrane complex
- CHAPS, 3-[(3-cholamidopropyl) dimethylammonio]-1-propanesulfonate
- DAMP, damage associated molecular pattern
- ECM, extracellular matrix
- HMECs, human microvascular endothelial cells
- NPCs, non-parenchymal cells
- PLECM, porcine-liver-derived extracellular matrix
- SDS, sodium dodecyl sulfate
- SEC, sinusoidal endothelial cell
- SEM, scanning electron microscopy
- biologic scaffold
- decellularization
- extracellular matrix
- liver tissue engineering
- organ engineering
Collapse
Affiliation(s)
- Denver M. Faulk
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Justin D. Wildemann
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA,Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA,Address for correspondence: Stephen F. Badylak, 450 Technology Drive, Suite 300, University of Pittsburgh, Pittsburgh, PA 15219, USA. Tel.: +412 624 5252; fax: +412 624 5256.
| |
Collapse
|
37
|
Carruthers CA, Dearth CL, Reing JE, Kramer CR, Gagne DH, Crapo PM, Garcia O, Badhwar A, Scott JR, Badylak SF. Histologic characterization of acellular dermal matrices in a porcine model of tissue expander breast reconstruction. Tissue Eng Part A 2014; 21:35-44. [PMID: 24941900 DOI: 10.1089/ten.tea.2014.0095] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acellular dermal matrices (ADMs) have been commonly used in expander-based breast reconstruction to provide inferolateral prosthesis coverage. Although the clinical performance of these biologic scaffold materials varies depending on a number of factors, an in-depth systematic characterization of the host response is yet to be performed. The present study evaluates the biochemical composition and structure of two ADMs, AlloDerm(®) Regenerative Tissue Matrix and AlloMax™ Surgical Graft, and provides a comprehensive spatiotemporal characterization in a porcine model of tissue expander breast reconstruction. METHODS Each ADM was characterized with regard to thickness, permeability, donor nucleic acid content, (residual double-stranded DNA [dsDNA]), and growth factors (basic fibroblast growth factor [bFGF], vascular endothelial growth factor [VEGF], and transforming growth factor-beta 1 [TGF-β1]). Cytocompatibility was evaluated by in vitro cell culture on the ADMs. The host response was evaluated at 4 and 12 weeks at various locations within the ADMs using established metrics of the inflammatory and tissue remodeling response: cell infiltration, multinucleate giant cell formation, extent of ADM remodeling, and neovascularization. RESULTS AlloMax incorporated more readily with surrounding host tissue as measured by earlier and greater cell infiltration, fewer foreign body giant cells, and faster remodeling of ADM. These findings correlated with the in vitro composition and cytocompatibility analysis, which showed AlloMax to more readily support in vitro cell growth. CONCLUSIONS AlloMax and AlloDerm demonstrated distinct remodeling characteristics in a porcine model of tissue expander breast reconstruction.
Collapse
|
38
|
Chung CG, James AW, Asatrian G, Chang L, Nguyen A, Le K, Bayani G, Lee R, Stoker D, Zhang X, Ting K, Péault B, Soo C. Human perivascular stem cell-based bone graft substitute induces rat spinal fusion. Stem Cells Transl Med 2014; 3:1231-41. [PMID: 25154782 DOI: 10.5966/sctm.2014-0027] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adipose tissue is an attractive source of mesenchymal stem cells (MSCs) because of its abundance and accessibility. We have previously defined a population of native MSCs termed perivascular stem cells (PSCs), purified from diverse human tissues, including adipose tissue. Human PSCs (hPSCs) are a bipartite cell population composed of pericytes (CD146+CD34-CD45-) and adventitial cells (CD146-CD34+CD45-), isolated by fluorescence-activated cell sorting and with properties identical to those of culture identified MSCs. Our previous studies showed that hPSCs exhibit improved bone formation compared with a sample-matched unpurified population (termed stromal vascular fraction); however, it is not known whether hPSCs would be efficacious in a spinal fusion model. To investigate, we evaluated the osteogenic potential of freshly sorted hPSCs without culture expansion and differentiation in a rat model of posterolateral lumbar spinal fusion. We compared increasing dosages of implanted hPSCs to assess for dose-dependent efficacy. All hPSC treatment groups induced successful spinal fusion, assessed by manual palpation and microcomputed tomography. Computerized biomechanical simulation (finite element analysis) further demonstrated bone fusion with hPSC treatment. Histological analyses showed robust endochondral ossification in hPSC-treated samples. Finally, we confirmed that implanted hPSCs indeed differentiated into osteoblasts and osteocytes; however, the majority of the new bone formation was of host origin. These results suggest that implanted hPSCs positively regulate bone formation via direct and paracrine mechanisms. In summary, hPSCs are a readily available MSC population that effectively forms bone without requirements for culture or predifferentiation. Thus, hPSC-based products show promise for future efforts in clinical bone regeneration and repair.
Collapse
Affiliation(s)
- Choon G Chung
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Greg Asatrian
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Le Chang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan Nguyen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Khoi Le
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Georgina Bayani
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David Stoker
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bruno Péault
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Chia Soo
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Department of Pathology and Laboratory Medicine, UCLA Operation Mend, and Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA; Marina Plastic Surgery Associates, Marina del Rey, California, USA; Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
39
|
Faulk DM, Londono R, Wolf MT, Ranallo CA, Carruthers CA, Wildemann JD, Dearth CL, Badylak SF. ECM hydrogel coating mitigates the chronic inflammatory response to polypropylene mesh. Biomaterials 2014; 35:8585-95. [PMID: 25043571 DOI: 10.1016/j.biomaterials.2014.06.057] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/29/2014] [Indexed: 12/20/2022]
Abstract
Polypropylene has been used as a surgical mesh material for several decades. This non-degradable synthetic polymer provides mechanical strength, a predictable host response, and its use has resulted in reduced recurrence rates for ventral hernia and pelvic organ prolapse. However, polypropylene and similar synthetic materials are associated with a chronic local tissue inflammatory response and dense fibrous tissue deposition. These outcomes have prompted variations in mesh design to minimize the surface area interface and increase integration with host tissue. In contrast, biologic scaffold materials composed of extracellular matrix (ECM) are rapidly degraded in-vivo and are associated with constructive tissue remodeling and minimal fibrosis. The objective of the present study was to assess the effects of an ECM hydrogel coating on the long-term host tissue response to polypropylene mesh in a rodent model of abdominal muscle injury. At 14 days post implantation, the ECM coated polypropylene mesh devices showed a decreased inflammatory response as characterized by the number and distribution of M1 macrophages (CD86+/CD68+) around mesh fibers when compared to the uncoated mesh devices. At 180 days the ECM coated polypropylene showed decreased density of collagen and amount of mature type I collagen deposited between mesh fibers when compared to the uncoated mesh devices. This study confirms and extends previous findings that an ECM coating mitigates the chronic inflammatory response and associated scar tissue deposition characteristic of polypropylene.
Collapse
Affiliation(s)
- Denver M Faulk
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Ricardo Londono
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; School of Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Matthew T Wolf
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Christian A Ranallo
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Christopher A Carruthers
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Justin D Wildemann
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Christopher L Dearth
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA
| | - Stephen F Badylak
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, USA.
| |
Collapse
|
40
|
Abstract
With advancements in biological and engineering sciences, the definition of an ideal biomaterial has evolved over the past 50 years from a substance that is inert to one that has select bioinductive properties and integrates well with adjacent host tissue. Biomaterials are a fundamental component of tissue engineering, which aims to replace diseased, damaged, or missing tissue with reconstructed functional tissue. Most biomaterials are less than satisfactory for pediatric patients because the scaffold must adapt to the growth and development of the surrounding tissues and organs over time. The pediatric community, therefore, provides a distinct challenge for the tissue engineering community.
Collapse
Affiliation(s)
- Timothy J Keane
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Bridgeside Point 2, 450 Technology Drive, Pittsburgh, Pennsylvania 15219; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Bridgeside Point 2, 450 Technology Drive, Pittsburgh, Pennsylvania 15219; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
41
|
Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T, Lazzari L, Soo C, Péault B. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci 2014; 71:1353-74. [PMID: 24158496 PMCID: PMC11113613 DOI: 10.1007/s00018-013-1462-6] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/17/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.
Collapse
Affiliation(s)
- Iain R. Murray
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Christopher C. West
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Winters R. Hardy
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
- Indiana Center for Vascular Biology and Medicine, Indianapolis, USA
| | - Aaron W. James
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tea Soon Park
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, USA
| | - Alan Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tulyapruek Tawonsawatruk
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lorenza Lazzari
- Cell Factory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Departments of Surgery and Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Bruno Péault
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
42
|
Crisan M, Corselli M, Chen WCW, Péault B. Perivascular cells for regenerative medicine. J Cell Mol Med 2014; 16:2851-60. [PMID: 22882758 PMCID: PMC4393715 DOI: 10.1111/j.1582-4934.2012.01617.x] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 08/02/2012] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are currently the best candidate therapeutic cells for regenerative medicine related to osteoarticular, muscular, vascular and inflammatory diseases, although these cells remain heterogeneous and necessitate a better biological characterization. We and others recently described that MSC originate from two types of perivascular cells, namely pericytes and adventitial cells and contain the in situ counterpart of MSC in developing and adult human organs, which can be prospectively purified using well defined cell surface markers. Pericytes encircle endothelial cells of capillaries and microvessels and express the adhesion molecule CD146 and the PDGFRβ, but lack endothelial and haematopoietic markers such as CD34, CD31, vWF (von Willebrand factor), the ligand for Ulex europaeus 1 (UEA1) and CD45 respectively. The proteoglycan NG2 is a pericyte marker exclusively associated with the arterial system. Besides its expression in smooth muscle cells, smooth muscle actin (αSMA) is also detected in subsets of pericytes. Adventitial cells surround the largest vessels and, opposite to pericytes, are not closely associated to endothelial cells. Adventitial cells express CD34 and lack αSMA and all endothelial and haematopoietic cell markers, as for pericytes. Altogether, pericytes and adventitial perivascular cells express in situ and in culture markers of MSC and display capacities to differentiate towards osteogenic, adipogenic and chondrogenic cell lineages. Importantly, adventitial cells can differentiate into pericyte-like cells under inductive conditions in vitro. Altogether, using purified perivascular cells instead of MSC may bring higher benefits to regenerative medicine, including the possibility, for the first time, to use these cells uncultured.
Collapse
Affiliation(s)
- Mihaela Crisan
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
43
|
Slivka PF, Dearth CL, Keane TJ, Meng FW, Medberry CJ, Riggio RT, Reing JE, Badylak SF. Fractionation of an ECM hydrogel into structural and soluble components reveals distinctive roles in regulating macrophage behavior. Biomater Sci 2014; 2:1521-34. [DOI: 10.1039/c4bm00189c] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Extracellular matrix (ECM) derived from mammalian tissues has been utilized to repair damaged or missing tissue and improve healing outcomes.
Collapse
Affiliation(s)
- P. F. Slivka
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
| | - C. L. Dearth
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
- Department of Surgery
- University of Pittsburgh
| | - T. J. Keane
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
- Department of Bioengineering
- University of Pittsburgh
| | - F. W. Meng
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
| | - C. J. Medberry
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
- Department of Bioengineering
- University of Pittsburgh
| | - R. T. Riggio
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
- Sanford School of Medicine
- University of South Dakota
| | - J. E. Reing
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
| | - S. F. Badylak
- McGowan Institute for Regenerative Medicine
- University of Pittsburgh
- Pittsburgh, USA
- Department of Surgery
- University of Pittsburgh
| |
Collapse
|
44
|
Turner NJ, Londono R, Dearth CL, Culiat CT, Badylak SF. Human NELL1 protein augments constructive tissue remodeling with biologic scaffolds. Cells Tissues Organs 2013; 198:249-65. [PMID: 24335144 DOI: 10.1159/000356491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
Biologic scaffolds composed of extracellular matrix (ECM) derived from decellularized tissues effectively reprogram key stages of the mammalian response to injury, altering the wound microenvironment from one that promotes scar tissue formation to one that stimulates constructive and functional tissue remodeling. In contrast, engineered scaffolds, composed of purified ECM components such as collagen, lack the complex ultrastructure and composition of intact ECM and may promote wound healing but lack factors that facilitate constructive and functional tissue remodeling. The objective of the present study was to test the hypothesis that addition of NELL1, a signaling protein that controls cell growth and differentiation, enhances the constructive tissue remodeling of a purified collagen scaffold. An abdominal wall defect model in the rat of 1.5-cm(2) partial thickness was used to compare the constructive remodeling of a bovine type I collagen scaffold to a biologic scaffold derived from small intestinal submucosa (SIS)-ECM with and without augmentation with 17 μg NELL1 protein. Samples were evaluated histologically at 14 days and 4 months. The contractile response of the defect site was also evaluated at 4 months. Addition of NELL1 protein improved the constructive remodeling of collagen scaffolds but not SIS-ECM scaffolds. Results showed an increase in the contractile force of the remodeled skeletal muscle and a fast:slow muscle composition similar to native tissue in the collagen-treated group. The already robust remodeling response to SIS-ECM was not enhanced by NELL1 at the dose tested. These findings suggest that NELL1 protein does contribute to the enhanced constructive remodeling of skeletal muscle.
Collapse
Affiliation(s)
- Neill J Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa., USA
| | | | | | | | | |
Collapse
|
45
|
Sicari BM, Dearth CL, Badylak SF. Tissue Engineering and Regenerative Medicine Approaches to Enhance the Functional Response to Skeletal Muscle Injury. Anat Rec (Hoboken) 2013; 297:51-64. [DOI: 10.1002/ar.22794] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Brian M. Sicari
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Cellular and Molecular Pathology Graduate Program; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Christopher L. Dearth
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| |
Collapse
|
46
|
Nieponice A, Ciotola FF, Nachman F, Jobe BA, Hoppo T, Londono R, Badylak S, Badaloni AE. Patch esophagoplasty: esophageal reconstruction using biologic scaffolds. Ann Thorac Surg 2013; 97:283-8. [PMID: 24266951 DOI: 10.1016/j.athoracsur.2013.08.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Standard techniques for surgical reconstruction of the esophagus remain suboptimal. Primary closure of diseased or injured esophagus has been associated with high morbidity, primarily due to leak and stricture, and synthetic materials are contraindicated due to the high risk of erosion and infection. Degradable bioscaffolds composed of extracellular matrix (ECM) have recently shown promising results in both pre-clinical and clinical settings to prevent stricture after extended endoscopic mucosal resection. We propose a novel surgical technique that utilizes an ECM scaffold as a reconstructive patch to augment the esophageal diameter during primary repair. METHODS Four patients requiring esophageal reconstruction underwent a patch esophagoplasty using an ECM scaffold composed of porcine urinary bladder ECM. The full thickness wall of the esophagus was replaced with an ECM patch that was sutured to the edges of the remaining esophagus, similar to the patch angioplasty performed in vascular procedures. RESULTS All patients had a favorable clinical outcome with immediate recovery from the procedure and reinstated oral intake after 7 days. One patient had a micro leak at day 5 that closed spontaneously 2 days after drainage. Follow-up studies including barium swallow and esophagogastroduodenoscopy (EGD) showed adequate esophageal emptying through the surgical segment in all patients. The EGD showed complete mucosal remodeling at 2 months, with approximately 20% area contraction at the patch level. The area of the defect was indistinguishable from surrounding healthy tissue. Biopsy of the patch area showed normal squamous epithelium. One of the patients had a separate intrathoracic stricture that required further surgery. Clinical outcomes were otherwise favorable in all cases. CONCLUSIONS An alternative for the treatment of esophageal stenosis is presented which uses a biological scaffold and an innovative surgical procedure. Additional work, including prospective studies and long-term follow-up, is required to fully evaluate the potential of this bioscaffold-based regenerative medicine approach for esophageal reconstruction.
Collapse
Affiliation(s)
- Alejandro Nieponice
- Esophageal Surgery Program, University of Favaloro, Buenos Aires, Argentina; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Franco F Ciotola
- Esophageal Surgery Program, University of Favaloro, Buenos Aires, Argentina
| | - Fabio Nachman
- Esophageal Surgery Program, University of Favaloro, Buenos Aires, Argentina
| | - Blair A Jobe
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Toshitaka Hoppo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ricardo Londono
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stephen Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adolfo E Badaloni
- Esophageal Surgery Program, University of Favaloro, Buenos Aires, Argentina
| |
Collapse
|
47
|
Crapo PM, Tottey S, Slivka PF, Badylak SF. Effects of biologic scaffolds on human stem cells and implications for CNS tissue engineering. Tissue Eng Part A 2013; 20:313-23. [PMID: 24004192 DOI: 10.1089/ten.tea.2013.0186] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Biologic scaffolds composed of mammalian extracellular matrix (ECM) promote constructive remodeling of tissues via mechanisms that include the recruitment of endogenous stem/progenitor cells, modulation of the host innate immune response, and influence of cell fate differentiation. Such scaffold materials are typically prepared by decellularization of source tissues and are prepared as sheets, powder, or hydrogels. It is plausible that ECM derived from an anatomically distinct tissue would have unique or specific effects on cells that naturally reside in this same tissue. The present study investigated the in vitro effect of a soluble form of ECM derived from central nervous system (CNS) tissue, specifically the spinal cord or brain, versus ECM derived from a non-CNS tissue; specifically, the urinary bladder on the behavior of neural stem cells (NSCs) and perivascular stem cells. All forms of ECM induce positive, mitogenic, and chemotactic effects at concentrations of approximately 100 μg/mL without affecting stem cell viability. CNS-derived ECMs also showed the ability to differentiate NSCs into neurons as indicted by βIII-tubulin expression in two-dimensional culture and neurite extension on the millimeter scale after 24 days of three-dimensional cultures in an ECM hydrogel. These results suggest that solubilized forms of ECM scaffold materials may facilitate the postinjury healing response in CNS tissues.
Collapse
Affiliation(s)
- Peter M Crapo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
48
|
Turner NJ, Keane TJ, Badylak SF. Lessons from developmental biology for regenerative medicine. ACTA ACUST UNITED AC 2013; 99:149-59. [DOI: 10.1002/bdrc.21040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Neill J. Turner
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Surgery, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Timothy J. Keane
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| |
Collapse
|
49
|
Cellular kinetics of perivascular MSC precursors. Stem Cells Int 2013; 2013:983059. [PMID: 24023546 PMCID: PMC3760099 DOI: 10.1155/2013/983059] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/13/2013] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and MSC-like multipotent stem/progenitor cells have been widely investigated for regenerative medicine and deemed promising in clinical applications. In order to further improve MSC-based stem cell therapeutics, it is important to understand the cellular kinetics and functional roles of MSCs in the dynamic regenerative processes. However, due to the heterogeneous nature of typical MSC cultures, their native identity and anatomical localization in the body have remained unclear, making it difficult to decipher the existence of distinct cell subsets within the MSC entity. Recent studies have shown that several blood-vessel-derived precursor cell populations, purified by flow cytometry from multiple human organs, give rise to bona fide MSCs, suggesting that the vasculature serves as a systemic reservoir of MSC-like stem/progenitor cells. Using individually purified MSC-like precursor cell subsets, we and other researchers have been able to investigate the differential phenotypes and regenerative capacities of these contributing cellular constituents in the MSC pool. In this review, we will discuss the identification and characterization of perivascular MSC precursors, including pericytes and adventitial cells, and focus on their cellular kinetics: cell adhesion, migration, engraftment, homing, and intercellular cross-talk during tissue repair and regeneration.
Collapse
|
50
|
Chen CW, Okada M, Proto JD, Gao X, Sekiya N, Beckman SA, Corselli M, Crisan M, Saparov A, Tobita K, Péault B, Huard J. Human pericytes for ischemic heart repair. Stem Cells 2013; 31:305-16. [PMID: 23165704 DOI: 10.1002/stem.1285] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 10/23/2012] [Indexed: 12/30/2022]
Abstract
Human microvascular pericytes (CD146(+)/34(-)/45(-)/56(-)) contain multipotent precursors and repair/regenerate defective tissues, notably skeletal muscle. However, their ability to repair the ischemic heart remains unknown. We investigated the therapeutic potential of human pericytes, purified from skeletal muscle, for treating ischemic heart disease and mediating associated repair mechanisms in mice. Echocardiography revealed that pericyte transplantation attenuated left ventricular dilatation and significantly improved cardiac contractility, superior to CD56+ myogenic progenitor transplantation, in acutely infarcted mouse hearts. Pericyte treatment substantially reduced myocardial fibrosis and significantly diminished infiltration of host inflammatory cells at the infarct site. Hypoxic pericyte-conditioned medium suppressed murine fibroblast proliferation and inhibited macrophage proliferation in vitro. High expression by pericytes of immunoregulatory molecules, including interleukin-6, leukemia inhibitory factor, cyclooxygenase-2, and heme oxygenase-1, was sustained under hypoxia, except for monocyte chemotactic protein-1. Host angiogenesis was significantly increased. Pericytes supported microvascular structures in vivo and formed capillary-like networks with/without endothelial cells in three-dimensional cocultures. Under hypoxia, pericytes dramatically increased expression of vascular endothelial growth factor-A, platelet-derived growth factor-β, transforming growth factor-β1 and corresponding receptors while expression of basic fibroblast growth factor, hepatocyte growth factor, epidermal growth factor, and angiopoietin-1 was repressed. The capacity of pericytes to differentiate into and/or fuse with cardiac cells was revealed by green fluorescence protein labeling, although to a minor extent. In conclusion, intramyocardial transplantation of purified human pericytes promotes functional and structural recovery, attributable to multiple mechanisms involving paracrine effects and cellular interactions.
Collapse
Affiliation(s)
- Chien-Wen Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|