1
|
Yang L, Xiao L, Gao W, Huang X, Wei F, Zhang Q, Xiao Y. Macrophages at Low-Inflammatory Status Improved Osteogenesis via Autophagy Regulation. Tissue Eng Part A 2024; 30:e766-e779. [PMID: 33678009 DOI: 10.1089/ten.tea.2021.0015] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence indicates that the interaction between immune and skeletal systems is vital in bone homeostasis. However, the detailed mechanisms between macrophage polarization and osteogenic differentiation of mesenchymal stromal cells (bone marrow-derived stromal cells [BMSCs]) remain largely unknown. We observed enhanced macrophage infiltration along with bone formation in vivo, which showed a transition from early-stage M1 phenotype to later stage M2 phenotype, cells at the transitional stage expressed both M1 and M2 markers that actively participated in osteogenesis, which was mimicked by stimulating macrophages with lower inflammatory stimulus (compared with typical M1). Using conditioned medium (CM) from M0, typical M1, low-inflammatory M1 (M1semi), and M2 macrophages, it was found that BMSCs treated with M1semi CM showed significantly induced migration, osteogenic differentiation, and mineralization, compared with others. Along with the induced osteogenesis, the autophagy level was the highest in M1semi CM-treated BMSCs, which was responsible for BMSC migration and osteogenic differentiation, as autophagy interruption significantly abolished this effect. This study indicated that low-inflammatory macrophages could activate autophagy in BMSCs to improve osteogenesis.
Collapse
Affiliation(s)
- Lan Yang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Lan Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Wendong Gao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Xin Huang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Fei Wei
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Qing Zhang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yin Xiao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
2
|
Marolt Presen D, Goeschl V, Hanetseder D, Ogrin L, Stetco AL, Tansek A, Pozenel L, Bruszel B, Mitulovic G, Oesterreicher J, Zipperle J, Schaedl B, Holnthoner W, Grillari J, Redl H. Prolonged cultivation enhances the stimulatory activity of hiPSC mesenchymal progenitor-derived conditioned medium. Stem Cell Res Ther 2024; 15:434. [PMID: 39551765 PMCID: PMC11572509 DOI: 10.1186/s13287-024-03960-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/25/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cells represent a scalable source of youthful tissue progenitors and secretomes for regenerative therapies. The aim of our study was to investigate the potential of conditioned medium (CM) from hiPSC-mesenchymal progenitors (hiPSC-MPs) to stimulate osteogenic differentiation of human bone marrow-derived mesenchymal stromal cells (MSCs). We also investigated whether prolonged cultivation or osteogenic pre-differentiation of hiPSC-MPs could enhance the stimulatory activity of CM. METHODS MSCs were isolated from 13 donors (age 20-90 years). CM derived from hiPSC-MPs was added to the MSC cultures and the effects on proliferation and osteogenic differentiation were examined after 14 days and 6 weeks. The stimulatory activity of hiPSC-MP-CM was compared with the activity of MSC-derived CM and with the activity of CM prepared from hiPSC-MPs pre-cultured in growth or osteogenic medium for 14 days. Comparative proteomic analysis of CM was performed to gain insight into the molecular components responsible for the stimulatory activity. RESULTS Primary bone marrow-derived MSC exhibited variability, with a tendency towards lower proliferation and tri-lineage differentiation in older donors. hiPSC-MP-CM increased the proliferation and alkaline phosphatase activity of MSC from several adult/aged donors after 14 days of continuous supplementation under osteogenic conditions. However, CM supplementation failed to improve the mineralization of MSC pellets after 6 weeks under osteogenic conditions. hiPSC-MP-CM showed greater enhancement of proliferation and ALP activity than CM derived from bone marrow-derived MSCs. Moreover, 14-day cultivation but not osteogenic pre-differentiation of hiPSC-MPs strongly enhanced CM stimulatory activity. Quantitative proteomic analysis of d14-CM revealed a distinct profile of components that formed a highly interconnected associations network with two clusters, one functionally associated with binding and organization of actin/cytoskeletal components and the other with structural constituents of the extracellular matrix, collagen, and growth factor binding. Several hub proteins were identified that were reported to have functions in cell-extracellular matrix interaction, osteogenic differentiation and development. CONCLUSIONS Our data show that hiPSC-MP-CM enhances early osteogenic differentiation of human bone marrow-derived MSCs and that prolonged cultivation of hiPSC-MPs enhances CM-stimulatory activity. Proteomic analysis of the upregulated protein components provides the basis for further optimization of hiPSC-MP-CM for bone regenerative therapies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria.
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria.
| | - Vanessa Goeschl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Dominik Hanetseder
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Laura Ogrin
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Alexandra-Larissa Stetco
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Anja Tansek
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Laura Pozenel
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Bella Bruszel
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, 3400, Austria
| | - Goran Mitulovic
- Clinical Department of Laboratory Medicine Proteomics Core Facility, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
- Bruker Austria, Lemböckgasse 47b, Vienna, 1230, Austria
| | - Johannes Oesterreicher
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Johannes Zipperle
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
- University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna, 1090, Austria
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Gregor-Mendel-Straße 33, Vienna, 1180, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| |
Collapse
|
3
|
Gonzalez-Fernandez P, Simula L, Jenni S, Jordan O, Allémann E. Hyaluronan-based hydrogel delivering glucose to mesenchymal stem cells intended to treat osteoarthritis. Int J Pharm 2024; 657:124139. [PMID: 38677396 DOI: 10.1016/j.ijpharm.2024.124139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Mesenchymal stem cell (MSC) therapy shows promise in regenerative medicine. For osteoarthritis (OA), MSCs delivered to the joint have a temporal window in which they can secrete growth factors and extracellular matrix molecules, contributing to cartilage regeneration and cell proliferation. However, upon injection in the non-vascularized joint, MSCs lacking energy supply, starve and die too quickly to efficiently deliver enough of these factors. To feed injected MSCs, we developed a hyaluronic acid (HA) derivative, where glucose is covalently bound to hyaluronic acid. To achieve this, the glucose moiety in 4-aminophenyl-β-D-glucopyranoside was linked to the HA backbone through amidation. The hydrogel was able to deliver glucose in a controlled manner using a trigger system based on hydrolysis catalyzed by endogenous ß-glucosidase. This led to glucose release from the hyaluronic acid backbone inside the cell. Indeed, our hydrogel proved to rescue starvation and cell mortality in a glucose-free medium. Our approach of adding a nutrient to the polymer backbone in hydrogels opens new avenues to deliver stem cells in poorly vascularized, nutrient-deficient environments, such as osteoarthritic joints, and for other regenerative therapies.
Collapse
Affiliation(s)
- Paula Gonzalez-Fernandez
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Luca Simula
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Sébastien Jenni
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
4
|
Wang Z, Chen X, Yan L, Wang W, Zheng P, Mohammadreza A, Liu Q. Antimicrobial peptides in bone regeneration: mechanism and potential. Expert Opin Biol Ther 2024; 24:285-304. [PMID: 38567503 DOI: 10.1080/14712598.2024.2337239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) are small-molecule peptides with a unique antimicrobial mechanism. Other notable biological activities of AMPs, including anti-inflammatory, angiogenesis, and bone formation effects, have recently received widespread attention. These remarkable bioactivities, combined with the unique antimicrobial mechanism of action of AMPs, have led to their increasingly important role in bone regeneration. AREAS COVERED In this review, on the one hand, we aimed to summarize information about the AMPs that are currently used for bone regeneration by reviewing published literature in the PubMed database. On the other hand, we also highlight some AMPs with potential roles in bone regeneration and their possible mechanisms of action. EXPERT OPINION The translation of AMPs to the clinic still faces many problems, but their unique antimicrobial mechanisms and other conspicuous biological activities suggest great potential. An in-depth understanding of the structure and mechanism of action of AMPs will help us to subsequently combine AMPs with different carrier systems and perform structural modifications to reduce toxicity and achieve stable release, which may be a key strategy for facilitating the translation of AMPs to the clinic.
Collapse
Affiliation(s)
- ZhiCheng Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - XiaoMan Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Liang Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - WenJie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - PeiJia Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Atashbahar Mohammadreza
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of International Education, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Denoeud C, Luo G, Paquet J, Boisselier J, Wosinski P, Moya A, Diallo A, Larochette N, Marinesco S, Meiller A, Becquart P, Moussi H, Vilquin JT, Logeart-Avramoglou D, Gand A, Larreta-Garde V, Pauthe E, Potier E, Petite H. Enzyme-controlled, nutritive hydrogel for mesenchymal stromal cell survival and paracrine functions. Commun Biol 2023; 6:1266. [PMID: 38092861 PMCID: PMC10719273 DOI: 10.1038/s42003-023-05643-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Abstract
Culture-adapted human mesenchymal stromal cells (hMSCs) are appealing candidates for regenerative medicine applications. However, these cells implanted in lesions as single cells or tissue constructs encounter an ischemic microenvironment responsible for their massive death post-transplantation, a major roadblock to successful clinical therapies. We hereby propose a paradigm shift for enhancing hMSC survival by designing, developing, and testing an enzyme-controlled, nutritive hydrogel with an inbuilt glucose delivery system for the first time. This hydrogel, composed of fibrin, starch (a polymer of glucose), and amyloglucosidase (AMG, an enzyme that hydrolyze glucose from starch), provides physiological glucose levels to fuel hMSCs via glycolysis. hMSCs loaded in these hydrogels and exposed to near anoxia (0.1% pO2) in vitro exhibited improved cell viability and angioinductive functions for up to 14 days. Most importantly, these nutritive hydrogels promoted hMSC viability and paracrine functions when implanted ectopically. Our findings suggest that local glucose delivery via the proposed nutritive hydrogel can be an efficient approach to improve hMSC-based therapeutic efficacy.
Collapse
Affiliation(s)
- Cyprien Denoeud
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Guotian Luo
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Joseph Paquet
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Julie Boisselier
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | | | - Adrien Moya
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Ahmad Diallo
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | | | | | - Anne Meiller
- Neuroscience Research Center, AniRA-NeuroChem Platform, Lyon, France
| | - Pierre Becquart
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Hilel Moussi
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Jean-Thomas Vilquin
- Sorbonne Université, INSERM, AIM, CNRS, Centre de Recherche en Myologie, Hôpital Pitié Salpêtrière, Paris, France
| | | | - Adeline Gand
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | | | - Emmanuel Pauthe
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | - Esther Potier
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Hervé Petite
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France.
| |
Collapse
|
6
|
Mahajan A, Bhattacharyya S. Immunomodulation by mesenchymal stem cells during osteogenic differentiation: Clinical implications during bone regeneration. Mol Immunol 2023; 164:143-152. [PMID: 38011783 DOI: 10.1016/j.molimm.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
Critical bone defects resulting in delayed and non-union are a major concern in the field of orthopedics. Over the past decade, mesenchymal stem cells (MSCs) have become a promising frontier for bone repair and regeneration owing to their high expansion rate and osteogenic differentiation potential ex vivo. MSCs have also long been associated with their ability to modulate immune response in the recipients. These can even skew the immune response towards pro-inflammatory or anti-inflammatory type by sensing their local microenvironment. MSCs adopt anti-inflammatory phenotype at bone injury site and secrete various immunomodulatory factors such as IDO, NO, TGFβ1 and PGE-2 which have redundant role in osteoblast differentiation and bone formation. As such, several studies have also sought to decipher the immunomodulatory effects of osteogenically differentiated MSCs. The present review discusses the immunomodulatory status of MSCs during their osteogenic differentiation and summarizes few mechanisms that cause immunosuppression by osteogenically differentiated MSCs and its implication during bone healing.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
7
|
Maroquenne M, Bourguignon M, Larochette N, El-Hafci H, Margottin M, Potier E, Logeart-Avramoglou D. The Lower in Vivo Osteogenicity of Adipose Tissue-Derived Stem Cells Correlates with a Higher Innate Immune Response. Stem Cell Rev Rep 2023; 19:2869-2885. [PMID: 37642900 DOI: 10.1007/s12015-023-10614-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Adipose tissue-derived mesenchymal stem cells (ATSCs) have been used as an alternative to bone marrow-derived mesenchymal stem cells (BMSCs) for bone tissue engineering applications. The ability of ATSCs to promote new bone formation remains lower than that of BMSCs. This study aimed to investigate the mechanisms underlying osteogenicity differences between human ATSCs and BMSCs in ceramic constructs, focusing on the effects of inflammation on this process. In contrast to ATSC-containing constructs, which did not induce bone formation in an ectopic mouse model, BMSC constructs consistently did so. Gene expression analysis revealed that human BMSCs, concomitantly with host murine progenitors, differentiated into the osteogenic lineage early post-implantation. In contrast, ATSCs differentiated later, when few implanted viable cells remained post-implantation, while the host murine cells did not differentiate. Comparison of the inflammatory profile in the cell constructs indicated concomitant upregulation of some human and murine inflammatory genes in the ATSC-constructs compared to the BMSC-constructs during the first-week post-implantation. The high level of chemokine production by the ATSCs was confirmed at the gene and protein levels before implantation. The immune cell recruitment within the constructs was then explored post-implantation. Higher numbers of TRAP-/ MRC1 (CD206) + multinucleated giant cells, NOS2 + M1, and ARG1 + M2 macrophages were present in the ATSC constructs than in the BMSC constructs. These results proved that ATSCs are a transient source of inflammatory cytokines promoting a transient immune response post-implantation; this milieu correlates with impaired osteogenic differentiation of both the implanted ATSCs and the host osteoprogenitor cells.
Collapse
Affiliation(s)
- Manon Maroquenne
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | | | | | - Hanane El-Hafci
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Morgane Margottin
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Esther Potier
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Delphine Logeart-Avramoglou
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France.
- Laboratoire de Biologie, Bioingénierie et Bioimagerie Ostéo-articulaires, Université Paris Cité, 10 Avenue de Verdun, Paris, F-75010, France.
| |
Collapse
|
8
|
Smolinská V, Boháč M, Danišovič Ľ. Current status of the applications of conditioned media derived from mesenchymal stem cells for regenerative medicine. Physiol Res 2023; 72:S233-S245. [PMID: 37888967 PMCID: PMC10669946 DOI: 10.33549/physiolres.935186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Recently published studies suggest that the paracrine substances released by mesenchymal stem cells (MSCs) are the primary motive behind the therapeutic action reported in these cells. Pre-clinical and clinical research on MSCs has produced promising outcomes. Furthermore, these cells are generally safe for therapeutic use and may be extracted from a variety of anatomical regions. Recent research has indicated, however, that transplanted cells do not live long and that the advantages of MSC treatment may be attributable to the large diversity of bioactive substances they create, which play a crucial role in the control of essential physiological processes. Secretome derivatives, such as conditioned media or exosomes, may provide significant benefits over cells in terms of manufacture, preservation, handling, longevity of the product, and potential as a ready-to-use biologic product. Despite their immunophenotypic similarities, the secretome of MSCs appears to vary greatly depending on the host's age and the niches in which the cells live. The secretome's effect on multiple biological processes such as angiogenesis, neurogenesis, tissue repair, immunomodulation, wound healing, anti-fibrotic, and anti-tumor for tissue maintenance and regeneration has been discovered. Defining the secretome of cultured cultivated MSC populations by conditioned media analysis will allow us to assess its potential as a novel treatment approach. This review will concentrate on accumulating data from pre-clinical and clinical trials pointing to the therapeutic value of the conditioned medium. At last, the necessity of characterizing the conditioned medium for determining its potential for cell-free treatment therapy will be emphasized in this study.
Collapse
|
9
|
Qiu G, Huang M, Ke D, Liu J, Weir MD, Ma T, Wang P, Oates TW, Schneider A, Xia Y, Xu HHK, Zhao L. Novel injectable calcium phosphate scaffold with human periodontal ligament stem cell encapsulation in microbeads for bone regeneration. FRONTIERS IN MATERIALS 2022; 9. [DOI: 10.3389/fmats.2022.977853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Objectives: 1) Develop a novel construct of human periodontal ligament stem cells (hPDLSCs) encapsulated in degradable alginate microbeads (DAMB) with human platelet lysate (hPL) and injectable calcium phosphate cement (ICPC); 2) Investigate the proliferation and osteogenic differentiation of hPDLSCs in ICPC with hPL as a xeno-free supplement and animal serum replacement for bone tissue engineering applications.Methods: hPDLSCs were encapsulated in alginate-fibrin microbeads (DAMB + fibrin), alginate-hPL degradable microbeads (DAMB + hPL), or alginate-fibrin-hPL microbeads (DAMB + fibrin + hPL). The proliferation and osteogenic differentiation of hPDLSCs were investigated in culturing with the ICPC scaffold.Results: Flexural strength of ICPC was 8.4 ± 0.91 MPa, and elastic modulus was 1.56 ± 0.1 GPa, exceeding those of cancellous bone. hPDLSCs had higher viability in DAMB + fibrin + hPL group than in DAMB + fibrin. ALP was 69.97 ± 16.96 mU/mg for ICPC + DAMB + fibrin + hPL group, higher than 30.68 ± 2.86 mU/mg of ICPC + DAMB + fibrin (p < 0.05) and 4.12 ± 1.65 mU/mg of control (p < 0.01). At 7 days, osteogenic gene expressions (ALP, RUNX2, COL1, and OPN) in ICPC + DAMB + fibrin + hPL and ICPC + DAMB + fibrin were 4–11 folds that of control. At 21 days, the hPDLSC-synthesized bone mineral amounts in ICPC + DAMB + fibrin + hPL and ICPC + DAMB + fibrin were 13.2 folds and 11.1 folds that of control group, respectively.Conclusion: The novel injectable CPC scaffold encapsulating hPDLSCs and hPL is promising to protect and deliver hPDLSCs. The hPL-based medium significantly enhanced the osteogenic differentiation of hPDLSCs in ICPC + DAMB + fibrin + hPL construct, suggesting a promising xeno-free approach for bone tissue regeneration applications.
Collapse
|
10
|
Kageyama T, Akieda H, Sonoyama Y, Sato K, Yoshikawa H, Isono H, Hirota M, Kitajima H, Chun YS, Maruo S, Fukuda J. Bone Beads Enveloped with Vascular Endothelial Cells for Bone Regenerative Medicine. Acta Biomater 2022:S1742-7061(22)00520-7. [PMID: 36030051 DOI: 10.1016/j.actbio.2022.08.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/27/2022]
Abstract
The transplantation of pre-vascularized bone grafts is a promising strategy to improve the efficacy of engraftment and bone regeneration. We propose a hydrogel microbead-based approach for preparing vascularized and high-density tissue grafts. Mesenchymal stem cell-encapsulated collagen microgels (2 µL), termed bone beads, were prepared through spontaneous constriction, which improved the density of the mesenchymal stem cells and collagen molecules by more than 15-fold from the initial day of culture. Constriction was attributed to cell-attractive forces and involved better osteogenic differentiation of mesenchymal stem cells than that of spheroids. This approach was scalable, and ∼2,000 bone beads were prepared semi-automatically using a liquid dispenser and spinner flask. The mechanical stimuli in the spinner flask further improved the osteogenic differentiation of the mesenchymal stem cells in the bone beads compared with that in static culture. Vascular endothelial cells readily attach to and cover the surface of bone beads. The in vitro assembly of the endothelial cell-enveloped bone beads resulted in microchannel formation in the interspaces between the bone beads. Significant effects of endothelialization on in vivo bone regeneration were shown in rats with cranial bone defects. The use of endothelialized bone beads may be a scalable and robust approach for treating large bone defects. STATEMENT OF SIGNIFICANCE: A unique aspect of this study is that the hMSC-encapsulated collagen microgels were prepared through spontaneous constriction, leading to the enrichment of collagen and cell density. This constriction resulted in favorable microenvironments for the osteogenic differentiation of hMSCs, which is superior to conventional spheroid culture. The microgel beads were then enveloped with vascular endothelial cells and assembled to fabricate a tissue graft with vasculature in the interspaces among the beads. The significant effects of endothelialization on in vivo bone regeneration were clearly demonstrated in rats with cranial bone defects. We believe that microgel beads covered with vascular endothelial cells provide a promising approach for engineering better tissue grafts for bone-regenerative medicine.
Collapse
Affiliation(s)
- Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN
| | - Hikaru Akieda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Yukie Sonoyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Ken Sato
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hiroshi Yoshikawa
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hitoshi Isono
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku Yokohama, Kanagawa 236-0004, JAPAN
| | - Makoto Hirota
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Hiroaki Kitajima
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Yang-Sook Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 110-799, KOREA
| | - Shoji Maruo
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN.
| |
Collapse
|
11
|
Brennan MÁ, Monahan DS, Brulin B, Gallinetti S, Humbert P, Tringides C, Canal C, Ginebra MP, Layrolle P. Biomimetic versus sintered macroporous calcium phosphate scaffolds enhanced bone regeneration and human mesenchymal stromal cell engraftment in calvarial defects. Acta Biomater 2021; 135:689-704. [PMID: 34520883 DOI: 10.1016/j.actbio.2021.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023]
Abstract
In contrast to sintered calcium phosphates (CaPs) commonly employed as scaffolds to deliver mesenchymal stromal cells (MSCs) targeting bone repair, low temperature setting conditions of calcium deficient hydroxyapatite (CDHA) yield biomimetic topology with high specific surface area. In this study, the healing capacity of CDHA administering MSCs to bone defects is evaluated for the first time and compared with sintered beta-tricalcium phosphate (β-TCP) constructs sharing the same interconnected macroporosity. Xeno-free expanded human bone marrow MSCs attached to the surface of the hydrophobic β-TCP constructs, while infiltrating the pores of the hydrophilic CDHA. Implantation of MSCs on CaPs for 8 weeks in calvaria defects of nude mice exhibited complete healing, with bone formation aligned along the periphery of β-TCP, and conversely distributed within the pores of CDHA. Human monocyte-osteoclast differentiation was inhibited in vitro by direct culture on CDHA compared to β-TCP biomaterials and indirectly by administration of MSC-conditioned media generated on CDHA, while MSCs increased osteoclastogenesis in both CaPs in vivo. MSC engraftment was significantly higher in CDHA constructs, and also correlated positively with bone in-growth in scaffolds. These findings demonstrate that biomimetic CDHA are favorable carriers for MSC therapies and should be explored further towards clinical bone regeneration strategies. STATEMENT OF SIGNIFICANCE: Delivery of mesenchymal stromal cells (MSCs) on calcium phosphate (CaP) biomaterials enhances reconstruction of bone defects. Traditional CaPs are produced at high temperature, but calcium deficient hydroxyapatite (CDHA) prepared at room temperature yields a surface structure more similar to native bone mineral. The objective of this study was to compare the capacity of biomimetic CDHA scaffolds with sintered β-TCP scaffolds for bone repair mediated by MSCs for the first time. In vitro, greater cell infiltration occurred in CDHA scaffolds and following 8 weeks in vivo, MSC engraftment was higher in CDHA compared to β-TCP, as was bone in-growth. These findings demonstrate the impact of material features such as surface structure, and highlight that CDHA should be explored towards clinical bone regeneration strategies.
Collapse
Affiliation(s)
- Meadhbh Á Brennan
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Biomedical Engineering, School of Engineering; and Regenerative Medicine Institute (REMEDI), School of Medicine, National University of Ireland (NUIG), Galway, Ireland
| | - David S Monahan
- Biomedical Engineering, School of Engineering; and Regenerative Medicine Institute (REMEDI), School of Medicine, National University of Ireland (NUIG), Galway, Ireland
| | - Bénédicte Brulin
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; INSERM, UMR 1214, ToNIC, CHU Purpan, Université Paul Sabatier, Toulouse 31024, France
| | - Sara Gallinetti
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Paul Humbert
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France
| | - Christina Tringides
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Harvard Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Cristina Canal
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Maria Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain; Institute of Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Baldiri i Reixach 10-12, Barcelona 08028, Spain
| | - Pierre Layrolle
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; INSERM, UMR 1214, ToNIC, CHU Purpan, Université Paul Sabatier, Toulouse 31024, France.
| |
Collapse
|
12
|
Humbert P, Brennan MÁ, De Lima J, Brion R, Adrait A, Charrier C, Brulin B, Trichet V, Couté Y, Blanchard F, Layrolle P. Apoptotic mesenchymal stromal cells support osteoclastogenesis while inhibiting multinucleated giant cells formation in vitro. Sci Rep 2021; 11:12144. [PMID: 34108508 PMCID: PMC8190145 DOI: 10.1038/s41598-021-91258-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022] Open
Abstract
In bone regeneration induced by the combination of mesenchymal stromal cells (MSCs) and calcium-phosphate (CaP) materials, osteoclasts emerge as a pivotal cell linking inflammation and bone formation. Favorable outcomes are observed despite short-term engraftments of implanted MSCs, highlighting their major paracrine function and the possible implication of cell death in modulating their secretions. In this work, we focused on the communication from MSCs towards osteoclasts-like cells in vitro. MSCs seeded on a CaP biomaterial or undergoing induced apoptosis produced a conditioned media favoring the development of osteoclasts from human CD14+ monocytes. On the contrary, MSCs’ apoptotic secretion inhibited the development of inflammatory multinucleated giant cells formed after IL-4 stimulation. Components of MSCs’ secretome before and after apoptotic stress were compared using mass spectrometry-based quantitative proteomics and a complementary immunoassay for major cytokines. CXCR-1 and CXCR-2 ligands, primarily IL-8/CXCL-8 but also the growth-regulated proteins CXCL-1, -2 or -3, were suggested as the major players of MSCs’ pro-osteoclastic effect. These findings support the hypothesis that osteoclasts are key players in bone regeneration and suggest that apoptosis plays an important role in MSCs’ effectiveness.
Collapse
Affiliation(s)
- Paul Humbert
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France.
| | - Meadhbh Á Brennan
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France.,Regenerative Medicine Institute, School of Medicine, and Bioengineering Department, School of Engineering, National University of Ireland, Galway, H91 TK33, Ireland
| | - Julien De Lima
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| | - Régis Brion
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France.,CHU Nantes, 44000, Nantes, France
| | - Annie Adrait
- Université Grenoble Alpes, CEA, INSERM, IRIG, BGE, 38000, Grenoble, France
| | - Céline Charrier
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| | - Bénédicte Brulin
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| | - Valérie Trichet
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| | - Yohann Couté
- Université Grenoble Alpes, CEA, INSERM, IRIG, BGE, 38000, Grenoble, France
| | - Frédéric Blanchard
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| | - Pierre Layrolle
- UMR 1238, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, INSERM, 44000, Nantes, France
| |
Collapse
|
13
|
Oral Bone Tissue Regeneration: Mesenchymal Stem Cells, Secretome, and Biomaterials. Int J Mol Sci 2021; 22:ijms22105236. [PMID: 34063438 PMCID: PMC8156243 DOI: 10.3390/ijms22105236] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, tissue engineering has become one of the most studied medical fields. Even if bone shows self-remodeling properties, in some cases, due to injuries or anomalies, bone regeneration can be required. In particular, oral bone regeneration is needed in the dentistry field, where the functional restoration of tissues near the tooth represents a limit for many dental implants. In this context, the application of biomaterials and mesenchymal stem cells (MSCs) appears promising for bone regeneration. This review focused on in vivo studies that evaluated bone regeneration using biomaterials with MSCs. Different biocompatible biomaterials were enriched with MSCs from different sources. These constructs showed an enhanced bone regenerative power in in vivo models. However, we discussed also a future perspective in tissue engineering using the MSC secretome, namely the conditioned medium and extracellular vesicles. This new approach has already shown promising results for bone tissue regeneration in experimental models.
Collapse
|
14
|
Zhang Y, Xie Y, Hao Z, Zhou P, Wang P, Fang S, Li L, Xu S, Xia Y. Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:18472-18487. [PMID: 33856781 DOI: 10.1021/acsami.0c22671] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Repair of large bone defects represents a major challenge for orthopedic surgeons. The newly formed microvessels inside grafts play a crucial role in successful bone tissue engineering. Previously, an active role for mesenchymal stem cell (MSC)-derived exosomes in blood vessel development and progression was suggested in the repair of multiple tissues. However, the reports on the application of MSC-derived exosomes in the repair of large bone defects are sparse. In this study, we encapsulated umbilical MSC-derived exosomes (uMSCEXOs) in hyaluronic acid hydrogel (HA-Gel) and combined them with customized nanohydroxyapatite/poly-ε-caprolactone (nHP) scaffolds to repair cranial defects in rats. Imaging and histological evaluation indicated that the uMSCEXOs/Gel/nHP composites markedly enhanced bone regeneration in vivo, and the uMSCEXOs might play a key role in this process. Moreover, the in vitro results demonstrated that uMSCEXOs promoted the proliferation, migration, and angiogenic differentiation of endothelial progenitor cells (EPCs) but did not significantly affect the osteogenic differentiation of BMSCs. Importantly, mechanistic studies revealed that exosomal miR-21 was the potential intercellular messenger that promoted angiogenesis by upregulating the NOTCH1/DLL4 pathway. In conclusion, our findings exhibit a promising exosome-based strategy in repairing large bone defects through enhanced angiogenesis, which potentially regulated by the miR-21/NOTCH1/DLL4 signaling axis.
Collapse
Affiliation(s)
- Yuntong Zhang
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yang Xie
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zichen Hao
- Department of Orthopaedics, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Panyu Zhou
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Panfeng Wang
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shuo Fang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lu Li
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shuogui Xu
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Xia
- Department of Emergency and Trauma, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
15
|
Ye D, Wu S, Zhang B, Hong C, Yang L. Characteristics and clinical potential of a cellularly modified gelatin sponge. J Appl Biomater Funct Mater 2021; 19:22808000211035061. [PMID: 34519565 DOI: 10.1177/22808000211035061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Human umbilical cord mesenchymal stem cells (HuMSCs) injected directly have been proven effective for improving chronic wounds. However, HuMSCs largely die within 14 days. The aim of study is to establish a cellularly modified gelatin sponge and investigate its characteristics and clinical potential. METHODS HuMSCs were isolated, expanded and seeded in a poly-L-lysine (PLL)-coated gelatin sponge. Fabricated gelatin sponges were estimated through observation of morphological surface and ultrastructure, following confirmed by histology method. Supernatants were collected at different times for enzyme-linked immunosorbent assays (ELISAs) to measure growth factors. The cell embedded gelatin sponges were implanted subcutaneously on the backs of mice and the samples were harvested and studied histologically. RESULTS HuMSCs gradually modified the gelatin sponge by depositing collagen and hyaluronic acid, and degrading the structure of gelatin, resulting in a dense, and elastic structure. Compared with cells cultured in monolayer, the levels of growth factors increased remarkably when HuMSCs were cultivated in the gelatin sponge. Upon subcutaneous implantation in the backs of mice, the cellularized gelatin sponges persisted for up to 2 months and eventually integrated into the host tissue, while blank gelatin sponges degraded completely by the end of the second month. CONCLUSION Gelatin sponge is a clinically accessible scaffold for HuMSCs implantation to maintain short-term survival of the cells and high-level production of growth factors, which demonstrates good clinical potential for enhancing wound healing.
Collapse
Affiliation(s)
- Danyan Ye
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Sixun Wu
- Department of Burns and Plastic Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Bingna Zhang
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Chuzhu Hong
- Clinical Research Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Lujun Yang
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
- Department of Burns and Plastic Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| |
Collapse
|
16
|
Jinnah AH, Whitlock P, Willey JS, Danelson K, Kerr BA, Hassan OA, Emory CL, Smith TL, Bracey DN. Improved osseointegration using porcine xenograft compared to demineralized bone matrix for the treatment of critical defects in a small animal model. Xenotransplantation 2020; 28:e12662. [PMID: 33242920 DOI: 10.1111/xen.12662] [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: 05/04/2020] [Revised: 10/04/2020] [Accepted: 11/09/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Autograft (AG) is the gold standard bone graft due to biocompatibility, osteoconductivity, osteogenicity, and osteoinductivity. Alternatives include allografts and xenografts (XG). METHODS We investigated the osseointegration and biocompatibility of a decellularized porcine XG within a critical defect animal model. We hypothesized that the XG will result in superior osseointegration compared to demineralized bone matrix (DBM) and equivalent immune response to AG. Critical defects were created in rat femurs and treated with XG, XG plus bone morphogenetic protein (BMP)-2, DBM, or AG. Interleukin (IL)-2 and IFN-gamma levels (inflammatory markers) were measured from animal blood draws at 1 week and 1 month post-operatively. At 1 month, samples underwent micro-positron-emission tomography (microPET) scans following 18-NaF injection. At 16 weeks, femurs were retrieved and sent for micro-computerized tomography (microCT) scans for blinded grading of osseointegration or were processed for histologic analysis with tartrate resistant acid phosphatase (TRAP) and pentachrome. RESULTS Enzyme linked immunosorbent assay testing demonstrated greater IL-2 levels in the XG vs. AG 1 week post-op; which normalized by 28 days post-op. MicroPET scans showed increased uptake within the AG compared to all groups. XG and XG + BMP-2 showed a trend toward increased uptake compared with DBM. MicroCT scans demonstrated increased osseointegration in XG and XG + BMP groups compared to DBM. Pentachrome staining demonstrated angiogenesis and endochondral bone formation. Furthermore, positive TRAP staining in samples from all groups indicated bone remodeling. CONCLUSIONS These data suggest that decellularized and oxidized porcine XG is biocompatible and at least equivalent to DBM in the treatment of a critical defect in a rat femur model.
Collapse
Affiliation(s)
- Alexander H Jinnah
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Patrick Whitlock
- Division of Pediatric Orthopaedics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jeffrey S Willey
- Department of Radiation/Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Kerry Danelson
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Bethany A Kerr
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Omer A Hassan
- Department of Pathology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Cynthia L Emory
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Thomas L Smith
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Daniel N Bracey
- Division of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| |
Collapse
|
17
|
Groeneveldt LC, Herpelinck T, Maréchal M, Politis C, van IJcken WFJ, Huylebroeck D, Geris L, Mulugeta E, Luyten FP. The Bone-Forming Properties of Periosteum-Derived Cells Differ Between Harvest Sites. Front Cell Dev Biol 2020; 8:554984. [PMID: 33324630 PMCID: PMC7723972 DOI: 10.3389/fcell.2020.554984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
The development of alternatives for autologous bone grafts is a major focus of bone tissue engineering. To produce living bone-forming implants, skeletal stem and progenitor cells (SSPCs) are envisioned as key ingredients. SSPCs can be obtained from different tissues including bone marrow, adipose tissue, dental pulp, and periosteum. Human periosteum-derived cells (hPDCs) exhibit progenitor cell characteristics and have well-documented in vivo bone formation potency. Here, we have characterized and compared hPDCs derived from tibia with craniofacial hPDCs, from maxilla and mandible, respectively, each representing a potential source for cell-based tissue engineered implants for craniofacial applications. Maxilla and mandible-derived hPDCs display similar growth curves as tibial hPDCs, with equal trilineage differentiation potential toward chondrogenic, osteogenic, and adipogenic cells. These craniofacial hPDCs are positive for SSPC-markers CD73, CD164, and Podoplanin (PDPN), and negative for CD146, hematopoietic and endothelial lineage markers. Bulk RNA-sequencing identified genes that are differentially expressed between the three sources of hPDC. In particular, differential expression was found for genes of the HOX and DLX family, for SOX9 and genes involved in skeletal system development. The in vivo bone formation, 8 weeks after ectopic implantation in nude mice, was observed in constructs seeded with tibial and mandibular hPDCs. Taken together, we provide evidence that hPDCs show different profiles and properties according to their anatomical origin, and that craniofacial hPDCs are potential sources for cell-based bone tissue engineering strategies. The mandible-derived hPDCs display - both in vitro and in vivo - chondrogenic and osteogenic differentiation potential, which supports their future testing for use in craniofacial bone regeneration applications.
Collapse
Affiliation(s)
- Lisanne C Groeneveldt
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,OMFS IMPATH Research Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tim Herpelinck
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Marina Maréchal
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Constantinus Politis
- OMFS IMPATH Research Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands.,Center for Biomics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA-R In Silico Medicine, Université de Liége, Liège, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Eskeatnaf Mulugeta
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
O'Connor K. A cautionary tale about the use of colony-forming efficiency as a proxy for the survival of mesenchymal stem cells. Stem Cell Res Ther 2020; 11:292. [PMID: 32678014 PMCID: PMC7367266 DOI: 10.1186/s13287-020-01805-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023] Open
Abstract
Colony-forming efficiency is a time-honored metric of the proliferation potential of mesenchymal stem cells (MSCs). This commentary raises a concern about the practice of using colony-forming efficiency as a proxy for cell survival. A recently published study from my laboratory investigated this issue. A marker of cellular aging, CD264, was employed to separate human bone marrow MSCs into populations of CD264− cells and culture-matched, aging CD264+ cells with high and low colony-forming efficiency, respectively. In vitro cell survival was evaluated with a single-cell assay; in vivo survival by bioluminescence imaging of MSCs attached to scaffolds that were implanted ectopically in immunodeficient mice. In our study, in vitro and in vivo survival of the MSC populations was independent of colony-forming efficiency. This finding indicates that caution should be exercised before using colony-forming efficiency as an indirect metric of cell survival. Direct measurement of survival may be required. Awareness of this issue should foster a robust experimental design and, thereby, facilitate the translation of MSC research into clinical practice.
Collapse
Affiliation(s)
- Kim O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, 6823 Saint Charles Ave., Boggs Center Room 300, New Orleans, LA, 70118, USA. .,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
19
|
Voskamp C, Koevoet WJLM, Somoza RA, Caplan AI, Lefebvre V, van Osch GJVM, Narcisi R. Enhanced Chondrogenic Capacity of Mesenchymal Stem Cells After TNFα Pre-treatment. Front Bioeng Biotechnol 2020; 8:658. [PMID: 32714905 PMCID: PMC7344141 DOI: 10.3389/fbioe.2020.00658] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/27/2020] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are promising cells to treat cartilage defects due to their chondrogenic differentiation potential. However, an inflammatory environment during differentiation, such as the presence of the cytokine TNFα, inhibits chondrogenesis and limits the clinical use of MSCs. On the other hand, it has been reported that exposure to TNFα during in vitro expansion can increase proliferation, migration, and the osteogenic capacity of MSCs and therefore can be beneficial for tissue regeneration. This indicates that the role of TNFα on MSCs may be dependent on the differentiation stage. To improve the chondrogenic capacity of MSCs in the presence of an inflamed environment, we aimed to determine the effect of TNFα on the chondrogenic differentiation capacity of MSCs. Here, we report that TNFα exposure during MSC expansion increased the chondrogenic differentiation capacity regardless of the presence of TNFα during chondrogenesis and that this effect of TNFα during expansion was reversed upon TNFα withdrawal. Interestingly, pre-treatment with another pro-inflammatory cytokine, IL-1β, did not increase the chondrogenic capacity of MSCs indicating that the pro-chondrogenic effect is specific for TNFα. Finally, we show that TNFα pre-treatment increased the levels of SOX11 and active β-catenin suggesting that these intracellular effectors may be useful targets to improve MSC-based cartilage repair. Overall, these results suggest that TNFα pre-treatment, by modulating SOX11 levels and WNT/β-catenin signaling, could be used as a strategy to improve MSC-based cartilage repair.
Collapse
Affiliation(s)
- Chantal Voskamp
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Wendy J. L. M. Koevoet
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Rodrigo A. Somoza
- Department of Biology and Skeletal Research Center, Case Western Reserve University, Cleveland, OH, United States
| | - Arnold I. Caplan
- Department of Biology and Skeletal Research Center, Case Western Reserve University, Cleveland, OH, United States
| | - Véronique Lefebvre
- Division of Orthopedic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Gerjo J. V. M. van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
20
|
Ranmuthu CDS, Ranmuthu CKI, Russell JC, Singhania D, Khan WS. Evaluating the Effect of Non-cellular Bioactive Glass-Containing Scaffolds on Osteogenesis and Angiogenesis in in vivo Animal Bone Defect Models. Front Bioeng Biotechnol 2020; 8:430. [PMID: 32478053 PMCID: PMC7240009 DOI: 10.3389/fbioe.2020.00430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The use of bone scaffolds to replace injured or diseased bone has many advantages over the currently used autologous and allogeneic options in clinical practice. This systematic review evaluates the current evidence for non-cellular scaffolds containing bioactive glass on osteogenesis and angiogenesis in animal bone defect models. Studies that reported results of osteogenesis via micro-CT and results of angiogenesis via Microfil perfusion or immunohistochemistry were included in the review. A literature search of PubMed, EMBASE and Scopus was carried out in November 2019 from which nine studies met the inclusion and exclusion criteria. Despite the significant heterogeneity in the composition of the scaffolds used in each study, it could be concluded that scaffolds containing bioactive glass improve bone regeneration in these models, both by osteogenic and angiogenic measures. Incorporation of additional elements into the glass network, using additives, and using biochemical factors generally had a beneficial effect. Comparing the different compositions of non-cellular bioactive glass containing scaffolds is however difficult due to the heterogeneity in bioactive glass compositions, fabrication methods and biochemical additives used.
Collapse
Affiliation(s)
| | | | - Jodie C. Russell
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Disha Singhania
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Wasim S. Khan
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Leppik L, Sielatycka K, Henrich D, Han Z, Wang H, Eischen-Loges MJ, Oliveira KMC, Bhavsar MB, Ratajczak MZ, Barker JH. Role of Adult Tissue-Derived Pluripotent Stem Cells in Bone Regeneration. Stem Cell Rev Rep 2019; 16:198-211. [PMID: 31828580 PMCID: PMC6987071 DOI: 10.1007/s12015-019-09943-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Background Bone marrow-derived mononuclear cells (BM-MNC) consist of a heterogeneous mix of mesenchymal stem cells (MSC), hematopoietic progenitor cells (HPC), endothelial progenitor cells (EPC), monocytes, lymphocytes and pluripotent stem cells. Whereas the importance of MSC and EPC has been well documented in bone healing and regeneration studies, the role of pluripotent stem cells is still poorly understood. In the present study we evaluated if and how Very Small Embryonic Like cells (VSEL), isolated from rat BM-MNC, contribute to bone healing. Methods Large bone defects were made in the femurs of 38 Sprague Dawley female rats and treated with β-TCP scaffold granules seeded with male VSEL; BM-MNC, VSEL-depleted BM-MNC or scaffold alone, and bone healing was evaluated at 8 weeks post-surgery. Results Bone healing was significantly increased in defects treated with VSEL and BM-MNC, compared to defects treated with VSEL-depleted BM-MNC. Donor cells were detected in new bone tissue, in all the defects treated with cells, and in fibrous tissue only in defects treated with VSEL-depleted BM-MNC. The number of CD68+ cells was the highest in the VSEL-depleted group, whereas the number of TRAP positive cells was the lowest in this group. Conclusions Based on the results, we can conclude that VSEL play a role in BM-MNC induced bone formation. In our rat femur defect model, in defects treated with VSEL-depleted BM-MNC, osteoclastogenesis and bone formation were decreased, and foreign body reaction was increased.
Collapse
Affiliation(s)
- Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany.
| | - K Sielatycka
- Institute of Biology, Faculty of Exact and Natural Science, University of Szczecin, Szczecin, Poland
| | - D Henrich
- Department of Trauma, Hand & Reconstructive Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - Z Han
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - H Wang
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - M J Eischen-Loges
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - K M C Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - M B Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - M Z Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - J H Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
22
|
Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal Stromal Cell-Based Bone Regeneration Therapies: From Cell Transplantation and Tissue Engineering to Therapeutic Secretomes and Extracellular Vesicles. Front Bioeng Biotechnol 2019; 7:352. [PMID: 31828066 PMCID: PMC6890555 DOI: 10.3389/fbioe.2019.00352] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Effective regeneration of bone defects often presents significant challenges, particularly in patients with decreased tissue regeneration capacity due to extensive trauma, disease, and/or advanced age. A number of studies have focused on enhancing bone regeneration by applying mesenchymal stromal cells (MSCs) or MSC-based bone tissue engineering strategies. However, translation of these approaches from basic research findings to clinical use has been hampered by the limited understanding of MSC therapeutic actions and complexities, as well as costs related to the manufacturing, regulatory approval, and clinical use of living cells and engineered tissues. More recently, a shift from the view of MSCs directly contributing to tissue regeneration toward appreciating MSCs as "cell factories" that secrete a variety of bioactive molecules and extracellular vesicles with trophic and immunomodulatory activities has steered research into new MSC-based, "cell-free" therapeutic modalities. The current review recapitulates recent developments, challenges, and future perspectives of these various MSC-based bone tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Traweger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Spinal Cord Injury & Tissue Regeneration Center Salzburg, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
23
|
Madsen SD, Jones SH, Tucker HA, Giler MK, Muller DC, Discher CT, Russell KC, Dobek GL, Sammarco MC, Bunnell BA, O'Connor KC. Survival of aging CD264 + and CD264 - populations of human bone marrow mesenchymal stem cells is independent of colony-forming efficiency. Biotechnol Bioeng 2019; 117:223-237. [PMID: 31612990 DOI: 10.1002/bit.27195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/20/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
In vivo mesenchymal stem cell (MSC) survival is relevant to therapeutic applications requiring engraftment and potentially to nonengraftment applications as well. MSCs are a mixture of progenitors at different stages of cellular aging, but the contribution of this heterogeneity to the survival of MSC implants is unknown. Here, we employ a biomarker of cellular aging, the decoy TRAIL receptor CD264, to compare the survival kinetics of two cell populations in human bone marrow MSC (hBM-MSC) cultures. Sorted CD264+ hBM-MSCs from two age-matched donors have elevated β-galactosidase activity, decreased differentiation potential and form in vitro colonies inefficiently relative to CD264- hBM-MSCs. Counterintuitive to their aging phenotype, CD264+ hBM-MSCs exhibited comparable survival to matched CD264- hBM-MSCs from the same culture during in vitro colony formation and in vivo when implanted ectopically in immunodeficient NIH III mice. In vitro and in vivo survival of these two cell populations were independent of colony-forming efficiency. These findings have ramifications for the preparation of hBM-MSC therapies given the prevalence of aging CD264+ cells in hBM-MSC cultures and the popularity of colony-forming efficiency as a quality control metric in preclinical and clinical studies with MSCs.
Collapse
Affiliation(s)
- Sean D Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Sean H Jones
- Department of Comparative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana
| | - H Alan Tucker
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Margaret K Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Dyllan C Muller
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| | - Carson T Discher
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| | - Katie C Russell
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| | - Georgina L Dobek
- Department of Comparative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Mimi C Sammarco
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana.,Department of Surgery, School of Medicine, Tulane University, New Orleans, Louisiana.,Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana.,Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Kim C O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, Louisiana.,Department of Surgery, School of Medicine, Tulane University, New Orleans, Louisiana.,Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
24
|
Bracey DN, Jinnah AH, Willey JS, Seyler TM, Hutchinson ID, Whitlock PW, Smith TL, Danelson KA, Emory CL, Kerr BA. Investigating the Osteoinductive Potential of a Decellularized Xenograft Bone Substitute. Cells Tissues Organs 2019; 207:97-113. [PMID: 31655811 PMCID: PMC6935535 DOI: 10.1159/000503280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Bone grafting is the second most common tissue transplantation procedure worldwide. One of the alternative methods for bone repair under investigation is a tissue-engineered bone substitute. An ideal property of tissue-engineered bone substitutes is osteoinductivity, defined as the ability to stimulate primitive cells to differentiate into a bone-forming lineage. In the current study, we use a decellularization and oxidation protocol to produce a porcine bone scaffold and examine whether it possesses osteoinductive potential and can be used to create a tissue-engineered bone microenvironment. The decellularization protocol was patented by our lab and consists of chemical decellularization and oxidation steps using combinations of deionized water, trypsin, antimicrobials, peracetic acid, and triton-X100. To test if the bone scaffold was a viable host, preosteoblasts were seeded and analyzed for markers of osteogenic differentiation. The osteoinductive potential was observed in vitro with similar osteogenic markers being expressed in preosteoblasts seeded on the scaffolds and demineralized bone matrix. To assess these properties in vivo, scaffolds with and without preosteoblasts preseeded were subcutaneously implanted in mice for 4 weeks. MicroCT scanning revealed 1.6-fold increased bone volume to total volume ratio and 1.4-fold increase in trabecular thickness in scaffolds after implantation. The histological analysis demonstrates new bone formation and blood vessel formation with pentachrome staining demonstrating osteogenesis and angiogenesis, respectively, within the scaffold. Furthermore, CD31+ staining confirmed the endothelial lining of the blood vessels. These results demonstrate that porcine bone maintains its osteoinductive properties after the application of a patented decellularization and oxidation protocol developed in our laboratory. Future work must be performed to definitively prove osteogenesis of human mesenchymal stem cells, biocompatibility in large animal models, and osteoinduction/osseointegration in a relevant clinical model in vivo. The ability to create a functional bone microenvironment using decellularized xenografts will impact regenerative medicine, orthopedic reconstruction, and could be used in the research of multiple diseases.
Collapse
Affiliation(s)
- Daniel N. Bracey
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Alexander H. Jinnah
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Jeffrey S. Willey
- Wake Forest Baptist Medical Center, Radiation Oncology, Winston Salem, NC, USA
| | | | | | | | - Thomas L. Smith
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Kerry A. Danelson
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Cynthia L. Emory
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Bethany A. Kerr
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
- Virginia Tech-Wake Forest University School for Bioengineering and Sciences, Winston Salem, NC, USA
- Wake Forest School of Medicine, Cancer Biology, Winston Salem, NC, USA
| |
Collapse
|
25
|
Curley M, Gonzalez ZN, Milne L, Hadoke P, Handel I, Péault B, Smith LB. Human Adipose-derived Pericytes Display Steroidogenic Lineage Potential in Vitro and Influence Leydig Cell Regeneration in Vivo in Rats. Sci Rep 2019; 9:15037. [PMID: 31636275 PMCID: PMC6803635 DOI: 10.1038/s41598-019-50855-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Exogenous androgen replacement is used to treat symptoms associated with low testosterone in males. However, adverse cardiovascular risk and negative fertility impacts impel development of alternative approaches to restore/maintain Leydig cell (LC) androgen production. Stem Leydig cell (SLC) transplantation shows promise in this regard however, practicality of SLC isolation/transplantation impede clinical translation. Multipotent human adipose-derived perivascular stem cells (hAd-PSCs) represent an attractive extragonadal stem cell source for regenerative therapies in the testis but their therapeutic potential in this context is unexplored. We asked whether hAd-PSCs could be converted into Leydig-like cells and determined their capacity to promote regeneration in LC-ablated rat testes. Exposure of hAd-PSCs to differentiation-inducing factors in vitro upregulated steroidogenic genes but did not fully induce LC differentiation. In vivo, no difference in LC-regeneration was noted between Sham and hAd-PSC-transplanted rats. Interestingly, Cyp17a1 expression increased in hAd-PSC-transplanted testes compared to intact vehicle controls and the luteinising hormone/testosterone ratio returned to Vehicle control levels which was not the case in EDS + Sham animals. Notably, hAd-PSCs were undetectable one-month after transplantation suggesting this effect is likely mediated via paracrine mechanisms during the initial stages of regeneration; either directly by interacting with regenerating LCs, or through indirect interactions with trophic macrophages.
Collapse
Affiliation(s)
- Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Zaniah N Gonzalez
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU, United Kingdom
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Patrick Hadoke
- The British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - Ian Handel
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, United Kingdom
| | - Bruno Péault
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU, United Kingdom.,Department of Orthopaedic Surgery and Broad Stem Cell Center, University of California at Los Angeles, 615 Charles E Young Dr S, Los Angeles, CA, 90095, USA
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom. .,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
26
|
The Effects of Hypoxia on the Immune-Modulatory Properties of Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:2509606. [PMID: 31687031 PMCID: PMC6800910 DOI: 10.1155/2019/2509606] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 08/11/2019] [Accepted: 09/09/2019] [Indexed: 01/09/2023] Open
Abstract
The therapeutic repertoire for life-threatening inflammatory conditions like sepsis, graft-versus-host reactions, or colitis is very limited in current clinical practice and, together with chronic ones, like the osteoarthritis, presents growing economic burden in developed countries. This urges the development of more efficient therapeutic modalities like the mesenchymal stem cell-based approaches. Despite the encouraging in vivo data, however, clinical trials delivered ambiguous results. Since one of the typical features of inflamed tissues is decreased oxygenation, the success of cellular therapy in inflammatory pathologies seems to be affected by the impact of oxygen depletion on transplanted cells. Here, we examine our current knowledge on the effect of hypoxia on the physiology of bone marrow-derived mesenchymal stromal cells, one of the most popular tools of practical cellular therapy, in the context of their immune-modulatory capacity.
Collapse
|
27
|
Humbert P, Brennan MÁ, Davison N, Rosset P, Trichet V, Blanchard F, Layrolle P. Immune Modulation by Transplanted Calcium Phosphate Biomaterials and Human Mesenchymal Stromal Cells in Bone Regeneration. Front Immunol 2019; 10:663. [PMID: 31001270 PMCID: PMC6455214 DOI: 10.3389/fimmu.2019.00663] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
A wide variety of biomaterials have been developed as both stabilizing structures for the injured bone and inducers of bone neoformation. They differ in chemical composition, shape, porosity, and mechanical properties. The most extensively employed and studied subset of bioceramics are calcium phosphate materials (CaPs). These materials, when transplanted alongside mesenchymal stem cells (MSCs), lead to ectopic (intramuscular and subcutaneous) and orthotopic bone formation in preclinical studies, and effective fracture healing in clinical trials. Human MSC transplantation in pre-clinical and clinical trials reveals very low engraftment in spite of successful clinical outcomes and their therapeutic actions are thought to be primarily through paracrine mechanisms. The beneficial role of transplanted MSC could rely on their strong immunomodulatory effect since, even without long-term engraftment, they have the ability to alter both the innate and adaptive immune response which is critical to facilitate new bone formation. This study presents the current knowledge of the immune response to the implantation of CaP biomaterials alone or in combination with MSC. In particular the central role of monocyte-derived cells, both macrophages and osteoclasts, in MSC-CaP mediated bone formation is emphasized. Biomaterial properties, such as macroporosity and surface microstructure, dictate the host response, and the ultimate bone healing cascade. Understanding intercellular communications throughout the inflammation, its resolution and the bone regeneration phase, is crucial to improve the current therapeutic strategies or develop new approaches.
Collapse
Affiliation(s)
- Paul Humbert
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | - Meadhbh Á. Brennan
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Noel Davison
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Instructure Labs, B.V., The Hague, Netherlands
| | - Philippe Rosset
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Centre Hospitalier Universitaire de Tours, Tours, France
| | - Valérie Trichet
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | | | - Pierre Layrolle
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| |
Collapse
|
28
|
Paudel S, Lee WH, Lee M, Zahoor T, Mitchell R, Yang SY, Zhao H, Schon L, Zhang Z. Intravenous administration of multipotent stromal cells and bone allograft modification to enhance allograft healing. Regen Med 2019; 14:199-211. [PMID: 30761943 DOI: 10.2217/rme-2018-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study investigated a coordinated strategy of revitalizing bone allograft with circulating multipotent stromal cells (MSCs). Materials & methods: After chemotactic and releasing assessments, stromal cell-derived factor 1 and platelet-derived growth factor BB in copolymers were coated on the bone allograft (AlloS-P). Allograft coated with copolymers alone (Allo), as controls, or AlloS-P was implanted into the femur of athymic mice, which received intravenous injections of human MSCs or saline at weeks 1, 2 and 3. Results: At week 8, the total callus volume (both cartilaginous and bony callus) around the allograft was the largest in the AlloS-P + MSC group (p < 0.05). Conclusion: Coating bone allograft with stromal cell-derived factor 1 and platelet-derived growth factor BB and intravenous injections of MSCs improved allograft incorporation.
Collapse
Affiliation(s)
- Sharada Paudel
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Wen-Han Lee
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Moses Lee
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Talal Zahoor
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Reed Mitchell
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Shang-You Yang
- Department of Orthopaedic Surgery, University of Kansas School of Medicine-Wichita, Wichita, KS, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Lew Schon
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Zijun Zhang
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| |
Collapse
|
29
|
The paracrine effects of human induced pluripotent stem cells promote bone-like structures via the upregulation of BMP expression in a mouse ectopic model. Sci Rep 2018; 8:17106. [PMID: 30459360 PMCID: PMC6244408 DOI: 10.1038/s41598-018-35546-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/26/2018] [Indexed: 01/20/2023] Open
Abstract
Use of human induced pluripotent stem cells (h-iPSCs) for bone tissue engineering is most appealing, because h-iPSCs are an inexhaustible source of osteocompetent cells. The present study investigated the contribution of undifferentiated h-iPSCs and elucidated aspects of the underlying mechanism(s) of the involvement of these cells to new bone formation. Implantation of undifferentiated h-iPSCs seeded on coral particles in ectopic sites of mice resulted in expression of osteocalcin and DMP-1, and in mineral content similar to that of the murine bone. The number of the implanted h-iPSCs decreased with time and disappeared by 30 days post-implantation. In contrast, expression of the murine osteogenic genes at day 15 and 30 post-implantation provided, for the first time, evidence that the implanted h-iPSCs affected the observed outcomes via paracrine mechanisms. Supporting evidence was provided because supernatant conditioned media from h-iPSCs (h-iPSC CM), promoted the osteogenic differentiation of human mesenchymal stem cells (h-MSCs) in vitro. Specifically, h-iPSC CM induced upregulation of the BMP-2, BMP-4 and BMP-6 genes, and promoted mineralization of the extracellular matrix. Given the current interest in the use of h-iPSCs for regenerative medicine applications, our study contributes new insights into aspects of the mechanism underlying the bone promoting capability of h-iPSCs.
Collapse
|
30
|
Maisani M, Ziane S, Ehret C, Levesque L, Siadous R, Le Meins J, Chevallier P, Barthélémy P, De Oliveira H, Amédée J, Mantovani D, Chassande O. A new composite hydrogel combining the biological properties of collagen with the mechanical properties of a supramolecular scaffold for bone tissue engineering. J Tissue Eng Regen Med 2017; 12:e1489-e1500. [DOI: 10.1002/term.2569] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/18/2017] [Accepted: 08/25/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Mathieu Maisani
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
- Lab. for Biomaterials & Bioengineering (CRC‐I), Dept. Min‐Met‐Materials Engineering & Research Center CHU de QuébecLaval University Québec City Canada
| | - Sophia Ziane
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| | - Camille Ehret
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| | - Lucie Levesque
- Lab. for Biomaterials & Bioengineering (CRC‐I), Dept. Min‐Met‐Materials Engineering & Research Center CHU de QuébecLaval University Québec City Canada
| | - Robin Siadous
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| | - Jean‐François Le Meins
- Laboratoire de Chimie des Polymères Organiques LCPO (UMR5629)‐Ecole Nationale Supérieure de Chimie, de Biologie et de Physique (ENSCBP)Institut Polytechnique de Bordeaux Talence France
| | - Pascale Chevallier
- Lab. for Biomaterials & Bioengineering (CRC‐I), Dept. Min‐Met‐Materials Engineering & Research Center CHU de QuébecLaval University Québec City Canada
| | | | - Hugo De Oliveira
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| | - Joëlle Amédée
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| | - Diego Mantovani
- Lab. for Biomaterials & Bioengineering (CRC‐I), Dept. Min‐Met‐Materials Engineering & Research Center CHU de QuébecLaval University Québec City Canada
| | - Olivier Chassande
- Laboratoire BIOTIS, Inserm U1026Université de Bordeaux Bordeaux France
| |
Collapse
|
31
|
Brennan MA, Renaud A, Guilloton F, Mebarki M, Trichet V, Sensebé L, Deschaseaux F, Chevallier N, Layrolle P. Inferior In Vivo Osteogenesis and Superior Angiogenesis of Human Adipose‐Derived Stem Cells Compared with Bone Marrow‐Derived Stem Cells Cultured in Xeno‐Free Conditions. Stem Cells Transl Med 2017; 6:2160-2172. [PMID: 29052365 PMCID: PMC5702520 DOI: 10.1002/sctm.17-0133] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
The possibility of using adipose tissue-derived stromal cells (ATSC) as alternatives to bone marrow-derived stromal cells (BMSC) for bone repair has garnered interest due to the accessibility, high cell yield, and rapid in vitro expansion of ATSC. For clinical relevance, their bone forming potential in comparison to BMSC must be proven. Distinct differences between ATSC and BMSC have been observed in vitro and comparison of osteogenic potential in vivo is not clear to date. The aim of the current study was to compare the osteogenesis of human xenofree-expanded ATSC and BMSC in vitro and in an ectopic nude mouse model of bone formation. Human MSC were implanted with biphasic calcium phosphate biomaterials in subcutis pockets for 8 weeks. Implant groups were: BMSC, ATSC, BMSC and ATSC mixed together in different ratios, as well as MSC primed with either osteogenic supplements (250 μM ascorbic acid, 10 mM β-glycerolphosphate, and 10 nM dexamethasone) or 50 ng/ml recombinant bone morphogenetic protein 4 prior to implantation. In vitro results show osteogenic gene expression and differentiation potentials of ATSC. Despite this, ATSC failed to form ectopic bone in vivo, in stark contrast to BMSC, although osteogenic priming did impart minor osteogenesis to ATSC. Neovascularization was enhanced by ATSC compared with BMSC; however, less ATSC engrafted into the implant compared with BMSC. Therefore, in the content of bone regeneration, the advantages of ATSC over BMSC including enhanced angiogenesis, may be negated by their lack of osteogenesis and prerequisite for osteogenic differentiation prior to transplantation. Stem Cells Translational Medicine 2017;6:2160-2172.
Collapse
Affiliation(s)
- Meadhbh A. Brennan
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Audrey Renaud
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Fabien Guilloton
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Miryam Mebarki
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Valerie Trichet
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Luc Sensebé
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Frederic Deschaseaux
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Nathalie Chevallier
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Pierre Layrolle
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| |
Collapse
|
32
|
Moya A, Larochette N, Bourguignon M, El-Hafci H, Potier E, Petite H, Logeart-Avramoglou D. Osteogenic potential of adipogenic predifferentiated human bone marrow-derived multipotent stromal cells for bone tissue-engineering. J Tissue Eng Regen Med 2017; 12:e1511-e1524. [PMID: 28875591 DOI: 10.1002/term.2571] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 07/13/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
Abstract
In the present study, we evaluated the benefits of an adipogenic predifferentiation, the pathway most closely related to osteoblastogenesis, on the pro-osteogenic potential of human adult multipotent bone marrow stromal cells (hBMSCs), both in vitro and in vivo. Adipogenic differentiation of hBMSCs for 14 days resulted in a heterogeneous cell population from which the most adipogenic-committed cells were eliminated by their lack of readhesion ability. Our results provided evidence that the select adherent adipogenic differentiated hBMSCs (sAD+ cells) express a gene profile characteristic of both adipogenic and osteogenic lineages. In vitro, when cultured in osteogenic medium, sAD+ differentiated along the osteogenic lineage faster than undifferentiated hBMSCs. In vivo, in an ectopic mouse model, sAD+ exhibited a significantly higher bone formation capability compared with undifferentiated hBMSCs. We sought, then, to investigate the underlying mechanisms responsible for such beneficial effects of adipogenic predifferentiation on bone formation and found that this outcome was not linked to a better cell survival post-implantation. The secretome of sAD+ was both proangiogenic and chemoattractant, but its potential did not supersede the one of undifferentiated hBMSCs. However, using co-culture systems, we observed that the sAD+ paracrine factors were pro-osteogenic on undifferentiated hBMSCs. In conclusion, adipogenic priming endows hBMSCs with high osteogenic potential as well as pro-osteogenic paracrine-mediated activity. This preconditioning appears as a promising strategy for bone tissue engineering technology in order to improve the hBMSC osteogenic potency in vivo.
Collapse
Affiliation(s)
- Adrien Moya
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | - Hanane El-Hafci
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Esther Potier
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Hervé Petite
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | |
Collapse
|
33
|
Maisani M, Pezzoli D, Chassande O, Mantovani D. Cellularizing hydrogel-based scaffolds to repair bone tissue: How to create a physiologically relevant micro-environment? J Tissue Eng 2017; 8:2041731417712073. [PMID: 28634532 PMCID: PMC5467968 DOI: 10.1177/2041731417712073] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022] Open
Abstract
Tissue engineering is a promising alternative to autografts or allografts for the regeneration of large bone defects. Cell-free biomaterials with different degrees of sophistication can be used for several therapeutic indications, to stimulate bone repair by the host tissue. However, when osteoprogenitors are not available in the damaged tissue, exogenous cells with an osteoblast differentiation potential must be provided. These cells should have the capacity to colonize the defect and to participate in the building of new bone tissue. To achieve this goal, cells must survive, remain in the defect site, eventually proliferate, and differentiate into mature osteoblasts. A critical issue for these engrafted cells is to be fed by oxygen and nutrients: the transient absence of a vascular network upon implantation is a major challenge for cells to survive in the site of implantation, and different strategies can be followed to promote cell survival under poor oxygen and nutrient supply and to promote rapid vascularization of the defect area. These strategies involve the use of scaffolds designed to create the appropriate micro-environment for cells to survive, proliferate, and differentiate in vitro and in vivo. Hydrogels are an eclectic class of materials that can be easily cellularized and provide effective, minimally invasive approaches to fill bone defects and favor bone tissue regeneration. Furthermore, by playing on their composition and processing, it is possible to obtain biocompatible systems with adequate chemical, biological, and mechanical properties. However, only a good combination of scaffold and cells, possibly with the aid of incorporated growth factors, can lead to successful results in bone regeneration. This review presents the strategies used to design cellularized hydrogel-based systems for bone regeneration, identifying the key parameters of the many different micro-environments created within hydrogels.
Collapse
Affiliation(s)
- Mathieu Maisani
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
- Laboratoire BioTis, Inserm U1026, Université de Bordeaux, Bordeaux, France
| | - Daniele Pezzoli
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
| | - Olivier Chassande
- Laboratoire BioTis, Inserm U1026, Université de Bordeaux, Bordeaux, France
| | - Diego Mantovani
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
| |
Collapse
|
34
|
Manassero M, Paquet J, Deschepper M, Viateau V, Retortillo J, Bensidhoum M, Logeart-Avramoglou D, Petite H. Comparison of Survival and Osteogenic Ability of Human Mesenchymal Stem Cells in Orthotopic and Ectopic Sites in Mice. Tissue Eng Part A 2016; 22:534-44. [PMID: 26896389 DOI: 10.1089/ten.tea.2015.0346] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Tissue constructs containing mesenchymal stem cells (MSCs) are appealing strategies for repairing large segmental bone defects, but they do not allow consistent bone healing and early cell death was identified as a cause of failure. However, little is known about cell survival in the clinical microenvironment encountered during bone healing process. Osteoconductive coral scaffold with or without luciferase-labeled human MSCs were implanted either in a critical segmental femoral bone defect stabilized by plate or subcutaneously in 44 mice. Cell survival was evaluated by serial bioluminescence imaging (BLI) and osteogenic capabilities by histology and microcomputed tomography. Comparisons between groups were performed with two-way analysis of variance test. Twenty mice were sacrificed 2 weeks after surgery for short-term evaluation and 24 mice at 10 weeks for long-term evaluation. BLI provided evidence of fast and continuous cell death: 85% decrease of the BLI signal over the first 2 weeks in both locations; in fact, less than 2% of the initial cell number was present in all constructs analyzed 4 weeks postimplantation and less than 1% of the initial cell number by 8 weeks postimplantation. By 2 weeks postimplantation, the amount of newly formed bone was self-limited and was similar to ectopic and orthotopic groups. By 10 weeks postimplantation, bone formation was significantly enhanced in the presence of MSCs in orthotopic site and the amount of newly formed bone in cell-containing constructs implanted in orthotopic locations was significantly higher than that observed in the ectopic group. Our results indicated that hMSCs promote bone formation despite early and massive cell death when loaded on coral scaffolds. Interestingly, bone formation was higher in orthotopic than ectopic site despite the same survival pattern. Ectopic implantation of cell-containing constructs is suitable to evaluate cell survival, but assessment of bone formation ability requires orthotopic implantation.
Collapse
Affiliation(s)
- Mathieu Manassero
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France .,2 Service de Chirurgie, Université Paris-Est , Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Joseph Paquet
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France
| | - Mickael Deschepper
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France
| | - Véronique Viateau
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France .,2 Service de Chirurgie, Université Paris-Est , Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jose Retortillo
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France .,2 Service de Chirurgie, Université Paris-Est , Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Morad Bensidhoum
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France
| | - Delphine Logeart-Avramoglou
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France
| | - Hervé Petite
- 1 Laboratoire de Bioingénierie et Bioimagerie Ostéo-Articulaires (B2OA-UMR CNRS 7052), Université Paris Diderot , Paris, France
| |
Collapse
|
35
|
Moya A, Larochette N, Paquet J, Deschepper M, Bensidhoum M, Izzo V, Kroemer G, Petite H, Logeart-Avramoglou D. Quiescence Preconditioned Human Multipotent Stromal Cells Adopt a Metabolic Profile Favorable for Enhanced Survival under Ischemia. Stem Cells 2016; 35:181-196. [PMID: 27578059 DOI: 10.1002/stem.2493] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/11/2016] [Accepted: 08/21/2016] [Indexed: 12/20/2022]
Abstract
A major impediment to the development of therapies with mesenchymal stem cells/multipotent stromal cells (MSC) is the poor survival and engraftment of MSCs at the site of injury. We hypothesized that lowering the energetic demand of MSCs by driving them into a quiescent state would enhance their survival under ischemic conditions. Human MSCs (hMSCs) were induced into quiescence by serum deprivation (SD) for 48 hours. Such preconditioned cells (SD-hMSCs) exhibited reduced nucleotide and protein syntheses compared to unpreconditioned hMSCs. SD-hMSCs sustained their viability and their ATP levels upon exposure to severe, continuous, near-anoxia (0.1% O2 ) and total glucose depletion for up to 14 consecutive days in vitro, as they maintained their hMSC multipotential capabilities upon reperfusion. Most importantly, SD-hMSCs showed enhanced viability in vivo for the first week postimplantation in mice. Quiescence preconditioning modified the energy-metabolic profile of hMSCs: it suppressed energy-sensing mTOR signaling, stimulated autophagy, promoted a shift in bioenergetic metabolism from oxidative phosphorylation to glycolysis and upregulated the expression of gluconeogenic enzymes, such as PEPCK. Since the presence of pyruvate in cell culture media was critical for SD-hMSC survival under ischemic conditions, we speculate that these cells may utilize some steps of gluconeogenesis to overcome metabolic stress. These findings support that SD preconditioning causes a protective metabolic adaptation that might be taken advantage of to improve hMSC survival in ischemic environments. Stem Cells 2017;35:181-196.
Collapse
Affiliation(s)
- Adrien Moya
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Nathanaël Larochette
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Joseph Paquet
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Mickael Deschepper
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Morad Bensidhoum
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Valentina Izzo
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,INSERM, U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,INSERM, U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital Q2:07, Stockholm, Sweden
| | - Hervé Petite
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Delphine Logeart-Avramoglou
- Laboratory of Bioengineering and Bioimaging for Osteo-Articular tissues, UMR 7052, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
36
|
Zamuner A, Cavo M, Scaglione S, Messina GML, Russo T, Gloria A, Marletta G, Dettin M. Design of Decorated Self-Assembling Peptide Hydrogels as Architecture for Mesenchymal Stem Cells. MATERIALS 2016; 9:ma9090727. [PMID: 28773852 PMCID: PMC5457046 DOI: 10.3390/ma9090727] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 01/09/2023]
Abstract
Hydrogels from self-assembling ionic complementary peptides have been receiving a lot of interest from the scientific community as mimetic of the extracellular matrix that can offer three-dimensional supports for cell growth or can become vehicles for the delivery of stem cells, drugs or bioactive proteins. In order to develop a 3D “architecture” for mesenchymal stem cells, we propose the introduction in the hydrogel of conjugates obtained by chemoselective ligation between a ionic-complementary self-assembling peptide (called EAK) and three different bioactive molecules: an adhesive sequence with 4 Glycine-Arginine-Glycine-Aspartic Acid-Serine-Proline (GRGDSP) motifs per chain, an adhesive peptide mapped on h-Vitronectin and the growth factor Insulin-like Growth Factor-1 (IGF-1). The mesenchymal stem cell adhesion assays showed a significant increase in adhesion and proliferation for the hydrogels decorated with each of the synthesized conjugates; moreover, such functionalized 3D hydrogels support cell spreading and elongation, validating the use of this class of self-assembly peptides-based material as very promising 3D model scaffolds for cell cultures, at variance of the less realistic 2D ones. Furthermore, small amplitude oscillatory shear tests showed that the presence of IGF-1-conjugate did not alter significantly the viscoelastic properties of the hydrogels even though differences were observed in the nanoscale structure of the scaffolds obtained by changing their composition, ranging from long, well-defined fibers for conjugates with adhesion sequences to the compact and dense film for the IGF-1-conjugate.
Collapse
Affiliation(s)
- Annj Zamuner
- Department of Industrial Engineering, University of Padua, Padua 35131, Italy.
| | - Marta Cavo
- National Research Council (CNR)-Institute of Electronics, Computer and Telecommunication Engineering, Genoa 16149, Italy.
- Department of Informatics, Bioengineering, Robotics and System Engineering (DIBRIS), University of Genoa, Genoa 16145, Italy.
| | - Silvia Scaglione
- National Research Council (CNR)-Institute of Electronics, Computer and Telecommunication Engineering, Genoa 16149, Italy.
| | - Grazia Maria Lucia Messina
- Laboratory for Molecular Surfaces and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Catania 95125, Italy.
| | - Teresa Russo
- Institute of Polymers, Composites and Biomaterials-CNR, Naples 80125, Italy.
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials-CNR, Naples 80125, Italy.
| | - Giovanni Marletta
- Laboratory for Molecular Surfaces and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Catania 95125, Italy.
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, Padua 35131, Italy.
| |
Collapse
|
37
|
Nuschke A, Rodrigues M, Rivera J, Yates C, Whaley D, Stolz D, Griffith L, Wells A. Epidermal Growth Factor Tethered to β-Tricalcium Phosphate Bone Scaffolds via a High-Affinity Binding Peptide Enhances Survival of Human Mesenchymal Stem Cells/Multipotent Stromal Cells in an Immune-Competent Parafascial Implantation Assay in Mice. Stem Cells Transl Med 2016; 5:1580-1586. [PMID: 27400798 PMCID: PMC5070502 DOI: 10.5966/sctm.2015-0326] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/08/2016] [Indexed: 12/25/2022] Open
Abstract
: Mesenchymal stem cells/multipotent stromal cells (MSCs) are attractive candidates for cell therapies owing to their ability to differentiate into many lineages. However, these cells often fail to survive when implanted into a harsh wound environment, limiting efficacy in vivo. To improve MSC survival, we previously found that tethered epidermal growth factor (tEGF) molecules that restrict epidermal growth factor receptor (EGFR) signaling to the cell surface provide resistance to death signals. To adapt this system to wound healing, we tethered epidermal growth factor (EGF) to tricalcium phosphate (TCP) particle scaffolds, clinically used in bone healing. Human primary MSCs seeded on TCP and mixed into a collagen-based gel were injected in the perifascial space of immunocompetent mice with or without tEGF attached to the surface. We found that tethering EGF to the TCP scaffolds yielded approximately a fourfold increase in MSC survival compared with non-EGF scaffolds at 21 days, as well as significant improvements in survival in the short term at 2 and 7 days after implantation. Overall, our approach to sustaining EGFR signaling reduced MSC death in vivo and may be useful for future cell therapies where MSCs typically die on implantation. SIGNIFICANCE Stem cells are limited as tissue replacements owing to rapid death induced in the hostile wound environment. It has been found that restricting epidermal growth factor (EGF) receptor signaling to the membrane provides a survival advantage. This report elucidates a method to tether EGF to bone induction material to improve the survival of mesenchymal stem cells/multipotent stromal cells in vivo.
Collapse
Affiliation(s)
- Austin Nuschke
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie Rodrigues
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jaime Rivera
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Cecelia Yates
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Health Promotion and Development, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania, USA
| | - Diana Whaley
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna Stolz
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
38
|
Allen AB, Butts EB, Copland IB, Stevens HY, Guldberg RE. Human platelet lysate supplementation of mesenchymal stromal cell delivery: issues of xenogenicity and species variability. J Tissue Eng Regen Med 2016; 11:2876-2884. [PMID: 27339032 DOI: 10.1002/term.2191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 02/03/2016] [Accepted: 03/14/2016] [Indexed: 01/10/2023]
Abstract
Immunogenicity of fetal bovine serum (FBS) poses a problem for its use in the propagation of autologous mesenchymal stromal cells (MSCs) for cell therapy. Human platelet lysate (hPL), an enriched growth factor solution containing mitogenic and angiogenic cues, has potential utility in replacing FBS for human MSC (hMSC) delivery strategies. Despite its potentiation of hMSC number in vitro, little is known concerning its capacity to supplement implanted hMSC-seeded constructs and promote tissue regeneration in vivo. In this study, we tested the effects of incorporating hPL in cell-seeded constructs implanted subcutaneously into immunocompromised rats, investigated in vitro interactions between hPL and rat MSCs (rMSCs) and determined interspecies variability in the PL product [hPL vs rat PL (rPL)] and its effect on cultured MSCs (hPL/hMSCs vs rPL/rMSCs). The overarching aim was to determine the utility of hPL to foster MSC survival in preclinical rodent models. Exposure to hPL-supplemented media resulted in rMSC death, by a process attributable to heat-labile proteins, but not membrane attack complex formation. In the in vitro syngeneic model, the rodent product proved fundamentally distinct from the human product, with rPL having substantially lower growth factor content than hPL. Moreover, contrary to the positive effects of hPL on hMSC expansion, rPL did not reduce rMSC doubling time for the serum concentrations examined. When tested in vivo, hPL did not improve cell survival within hydrogel constructs through 2 weeks postimplantation. In summary, this study highlights the many facets of xenogenicity and interspecies variability that must be considered in the preclinical evaluation of hPL. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ashley B Allen
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Emily B Butts
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ian B Copland
- Department of Haematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hazel Y Stevens
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert E Guldberg
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
39
|
Ahn J, Kumar H, Cha BH, Park S, Arai Y, Han I, Park SG, Lee SH. AIMP1 downregulation restores chondrogenic characteristics of dedifferentiated/degenerated chondrocytes by enhancing TGF-β signal. Cell Death Dis 2016; 7:e2099. [PMID: 26890138 PMCID: PMC5399188 DOI: 10.1038/cddis.2016.17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 12/11/2022]
Abstract
Dedifferentiation and degeneration of chondrocytes critically influences the efficiency of cartilage repair. One of the causes is the defect of transforming growth factor (TGF)-β signaling that promotes chondrogenic differentiation and degeneration. In the present study, we found that aminoacyl-tRNA synthetase-interacting multifunctional protein 1 (AIMP1) negatively regulates TGF-β signaling via interactions with Smad2 and Smad3 in immunoprecipitation assay and luciferase assay. In addition, we observed that the AIMP1 expression level was significantly increased in osteoarthritis (OA) patient-derived degenerated chondrocytes compared with healthy control. So, we hypothesized that downregulation of AIMP1 using small-interfering RNA (siRNA) technology in dedifferentiated (collected at passage #6) and degenerated (obtained from OA-affected areas) chondrocytes could lead to recover TGF-β signaling in both chondrocytes. Indeed, AIMP1 downregulation restored TGF-β signaling by promoting phosphorylation of Smad2 and Smad3, which shows redifferentiated characteristics in both dedifferentiated and degenerated chondrocytes. Additionally, implantation analyses using in vivo mouse model clearly showed that AIMP1 downregulation resulted in the increased chondrogenic potential as well as the enhanced cartilage tissue formation in both dedifferentiated and degenerated chondrocytes. Histological analyses clarified that AIMP1 downregulation increased expression levels of collagen type II (Col II) and aggrecan, but not Col I expression. Taken together, these data indicate that AIMP1 downregulation using siRNA is a novel tool to restore TGF-β signaling and thereby increases the chondrogenic potential of dedifferentiated/degenerated chondrocytes, which could be further developed as a therapeutic siRNA to treat OA.
Collapse
Affiliation(s)
- J Ahn
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - H Kumar
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea.,Department of Neurosurgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - B-H Cha
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - S Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Y Arai
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - I Han
- Department of Neurosurgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - S G Park
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, Republic of Korea
| | - S-H Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
40
|
Abstract
Bone is one of the few tissues to display a true potential for regeneration. Fracture healing is an obvious example where regeneration occurs through tightly regulated sequences of molecular and cellular events which recapitulate tissue formation seen during embryogenesis. Still in some instances, bone regeneration does not occur properly (i.e. critical size lesions) and an appropriate therapeutic intervention is necessary. Successful replacement of bone by tissue engineering will likely depend on the recapitulation of this flow of events. In fact, bone regeneration requires cross-talk between microenvironmental factors and cells; for example, resident mesenchymal progenitors are recruited and properly guided by soluble and insoluble signaling molecules. Tissue engineering attempts to reproduce and to mimic this natural milieu by delivering cells capable of differentiating into osteoblasts, inducing growth factors and biomaterials to support cellular attachment, proliferation, migration, and matrix deposition. In the last two decades, a significant effort has been made by the scientific community in the development of methods and protocols to repair and regenerate tissues such as bone, cartilage, tendons, and ligaments. In this same period, great advancements have been achieved in the biology of stem cells and on the mechanisms governing "stemness". Unfortunately, after two decades, effective clinical translation does not exist, besides a few limited examples. Many years have passed since cell-based regenerative therapies were first described as "promising approaches", but this definition still engulfs the present literature. Failure to envisage translational cell therapy applications in routine medical practice evidences the existence of unresolved scientific and technical struggles, some of which still puzzle researchers in the field and are presented in this chapter.
Collapse
Affiliation(s)
- Rodolfo Quarto
- Stem Cell Laboratory, Department of Experimental Medicine, University of Genova, c/o Advanced Biotechnology Center, L.go R. Benzi, 10, 16132, Genoa, Italy.
| | - Paolo Giannoni
- Stem Cell Laboratory, Department of Experimental Medicine, University of Genova, c/o Advanced Biotechnology Center, L.go R. Benzi, 10, 16132, Genoa, Italy
| |
Collapse
|
41
|
Tethering of Epidermal Growth Factor (EGF) to Beta Tricalcium Phosphate (βTCP) via Fusion to a High Affinity, Multimeric βTCP-Binding Peptide: Effects on Human Multipotent Stromal Cells/Connective Tissue Progenitors. PLoS One 2015; 10:e0129600. [PMID: 26121597 PMCID: PMC4488278 DOI: 10.1371/journal.pone.0129600] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/11/2015] [Indexed: 12/14/2022] Open
Abstract
Transplantation of freshly-aspirated autologous bone marrow, together with a scaffold, is a promising clinical alternative to harvest and transplantation of autologous bone for treatment of large defects. However, survival proliferation, and osteogenic differentiation of the marrow-resident stem and progenitor cells with osteogenic potential can be limited in large defects by the inflammatory microenvironment. Previous studies using EGF tethered to synthetic polymer substrates have demonstrated that surface-tethered EGF can protect human bone marrow-derived osteogenic stem and progenitor cells from pro-death inflammatory cues and enhance their proliferation without detriment to subsequent osteogenic differentiation. The objective of this study was to identify a facile means of tethering EGF to clinically-relevant βTCP scaffolds and to demonstrate the bioactivity of EGF tethered to βTCP using stimulation of the proliferative response of human bone-marrow derived mesenchymal stem cells (hBMSC) as a phenotypic metric. We used a phage display library and panned against βTCP and composites of βTCP with a degradable polyester biomaterial, together with orthogonal blocking schemes, to identify a 12-amino acid consensus binding peptide sequence, LLADTTHHRPWT, with high affinity for βTCP. When a single copy of this βTCP-binding peptide sequence was fused to EGF via a flexible peptide tether domain and expressed recombinantly in E. coli together with a maltose-binding domain to aid purification, the resulting fusion protein exhibited modest affinity for βTCP. However, a fusion protein containing a linear concatamer containing 10 repeats of the binding motif the resulting fusion protein showed high affinity stable binding to βTCP, with only 25% of the protein released after 7 days at 37oC. The fusion protein was bioactive, as assessed by its abilities to activate kinase signaling pathways downstream of the EGF receptor when presented in soluble form, and to enhance the proliferation of hBMSC when presented in tethered form on commercial βTCP bone regeneration scaffolds.
Collapse
|
42
|
Todeschi MR, El Backly R, Capelli C, Daga A, Patrone E, Introna M, Cancedda R, Mastrogiacomo M. Transplanted Umbilical Cord Mesenchymal Stem Cells Modify the In Vivo Microenvironment Enhancing Angiogenesis and Leading to Bone Regeneration. Stem Cells Dev 2015; 24:1570-81. [PMID: 25685989 DOI: 10.1089/scd.2014.0490] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Umbilical cord mesenchymal stem cells (UC-MSCs) show properties similar to bone marrow mesenchymal stem cells (BM-MSCs), although controversial data exist regarding their osteogenic potential. We prepared clinical-grade UC-MSCs from Wharton's Jelly and we investigated if UC-MSCs could be used as substitutes for BM-MSCs in muscoloskeletal regeneration as a more readily available and functional source of MSCs. UC-MSCs were loaded onto scaffolds and implanted subcutaneously (ectopically) and in critical-sized calvarial defects (orthotopically) in mice. For live cell-tracking experiments, UC-MSCs were first transduced with the luciferase gene. Angiogenic properties of UC-MSCs were tested using the mouse metatarsal angiogenesis assay. Cell secretomes were screened for the presence of various cytokines using an array assay. Analysis of implanted scaffolds showed that UC-MSCs, contrary to BM-MSCs, remained detectable in the implants for 3 weeks at most and did not induce bone formation in an ectopic location. Instead, they induced a significant increase of blood vessel ingrowth. In agreement with these observations, UC-MSC-conditioned medium presented a distinct and stronger proinflammatory/chemotactic cytokine profile than BM-MSCs and a significantly enhanced angiogenic activity. When UC-MSCs were orthotopically transplanted in a calvarial defect, they promoted increased bone formation as well as BM-MSCs. However, at variance with BM-MSCs, the new bone was deposited through the activity of stimulated host cells, highlighting the importance of the microenvironment on determining cell commitment and response. Therefore, we propose, as therapy for bone lesions, the use of allogeneic UC-MSCs by not depositing bone matrix directly, but acting through the activation of endogenous repair mechanisms.
Collapse
Affiliation(s)
- Maria Rosa Todeschi
- 1 Department of Experimental Medicine (DIMES), University of Genoa , Genoa, Italy
| | - Rania El Backly
- 1 Department of Experimental Medicine (DIMES), University of Genoa , Genoa, Italy .,2 Faculty of Dentistry, Alexandria University , Alexandria, Egypt
| | - Chiara Capelli
- 3 A.O.Papa Giovanni XXIII-USS Center of Cell Therapy "G. Lanzani" USC Hematology , Bergamo, Italy
| | - Antonio Daga
- 4 IRCCS AOU San Martino-IST National Cancer Research Institute , Genoa, Italy
| | - Eugenio Patrone
- 5 Department of Prosthetic Dentistry, University of Genoa , Genoa, Italy
| | - Martino Introna
- 3 A.O.Papa Giovanni XXIII-USS Center of Cell Therapy "G. Lanzani" USC Hematology , Bergamo, Italy
| | - Ranieri Cancedda
- 1 Department of Experimental Medicine (DIMES), University of Genoa , Genoa, Italy .,4 IRCCS AOU San Martino-IST National Cancer Research Institute , Genoa, Italy
| | - Maddalena Mastrogiacomo
- 1 Department of Experimental Medicine (DIMES), University of Genoa , Genoa, Italy .,4 IRCCS AOU San Martino-IST National Cancer Research Institute , Genoa, Italy
| |
Collapse
|
43
|
Stegen S, van Gastel N, Carmeliet G. Bringing new life to damaged bone: the importance of angiogenesis in bone repair and regeneration. Bone 2015; 70:19-27. [PMID: 25263520 DOI: 10.1016/j.bone.2014.09.017] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/26/2022]
Abstract
Bone has the unique capacity to heal without the formation of a fibrous scar, likely because several of the cellular and molecular processes governing bone healing recapitulate the events during skeletal development. A critical component in bone healing is the timely appearance of blood vessels in the fracture callus. Angiogenesis, the formation of new blood vessels from pre-existing ones, is stimulated after fracture by the local production of numerous angiogenic growth factors. The fracture vasculature not only supplies oxygen and nutrients, but also stem cells able to differentiate into osteoblasts and in a later phase also the ions necessary for mineralization. This review provides a concise report of the regulation of angiogenesis by bone cells, its importance during bone healing and its possible therapeutic applications in bone tissue engineering. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
44
|
Seebach E, Freischmidt H, Holschbach J, Fellenberg J, Richter W. Mesenchymal stroma cells trigger early attraction of M1 macrophages and endothelial cells into fibrin hydrogels, stimulating long bone healing without long-term engraftment. Acta Biomater 2014; 10:4730-4741. [PMID: 25058402 DOI: 10.1016/j.actbio.2014.07.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/10/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Implantation of mesenchymal stroma cells (MSCs) is an attractive approach to stimulate closure of large bone defects but an optimal carrier has yet to be defined. MSCs may display trophic and/or immunomodulatory features or stimulate bone healing by their osteogenic activity. The aim of this study was to unravel whether fibrin hydrogel supports early actions of implanted MSCs, such as host cell recruitment, immunomodulation and tissue regeneration, in long bone defects. Female rats received cell-free fibrin or male MSCs embedded in a fibrin carrier into plate-stabilized femoral bone defects. Removed callus was analyzed for host cell invasion (day 6), local cytokine expression (days 3 and 6) and persistence of male MSCs (days 3, 6, 14 and 28). Fibrin-MSC composites triggered fast attraction of host cells into the hydrogel while cell-free fibrin implants were not invaded. A migration front dominated by M1 macrophages and endothelial progenitor cells formed while M2 macrophages remained sparse. Only MSC-seeded fibrin hydrogel stimulated early tissue maturation and primitive vessel formation at day 6 in line with significantly higher VEGF mRNA levels recorded at day 3. Local TNF-α, IL-1β and IL-10 expression indicated a balanced immune cell activity independent of MSC implantation. Implanted MSCs persisted until day 14 but not day 28. Our results demonstrate that fibrin hydrogel is an attractive carrier for MSC implantation into long bone defects, supporting host cell attraction and pro-angiogenic activity. By this angiogenesis, implant integration and tissue maturation was stimulated in long bone healing independent of long-term engraftment of implanted MSCs.
Collapse
Affiliation(s)
- Elisabeth Seebach
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany
| | - Holger Freischmidt
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany
| | - Jeannine Holschbach
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany
| | - Jörg Fellenberg
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| |
Collapse
|
45
|
Brennan MÁ, Renaud A, Amiaud J, Rojewski MT, Schrezenmeier H, Heymann D, Trichet V, Layrolle P. Pre-clinical studies of bone regeneration with human bone marrow stromal cells and biphasic calcium phosphate. Stem Cell Res Ther 2014; 5:114. [PMID: 25311054 PMCID: PMC4445278 DOI: 10.1186/scrt504] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 09/30/2014] [Indexed: 02/15/2023] Open
Abstract
INTRODUCTION Repair of large bone defects remains a significant clinical challenge. Bone marrow stromal cells (BMSCs), a subset of which is known as bone marrow-derived mesenchymal stem cells, show therapeutic potential for bone regeneration. However, their isolation, expansion and implantation will need to be conducted under good manufacturing practices (GMP) at separate locations. An investigation which mimics this clinical scenario where large bone defects shall be regenerated is required before clinical trials can be initiated. METHODS Seven batches of 100 million human ex-vivo expanded BMSCs from five donors were transported fresh in syringes from a GMP facility in Germany to France. BMSCs were mixed with biphasic calcium phosphate (BCP) biomaterial prior to subcutaneous implantation in nude mice. The capacity of BMSCs in unison with BCP to regenerate critical sized cranial bone defects was also evaluated. BMSCs expressing luciferase were used to assess the viability and bio-distribution of implanted cells. In situ hybridization, using the human-specific repetitive Alu sequence, was performed for the identification of human cells in explants. RESULTS Eight weeks after implantation of BMSCs, mineralized bone containing mature bone marrow territories was formed in ectopic sites and in calvaria defects. Significant loss of cell viability was observed by bioluminescence imaging and only 1.5 percent of the initial number of transplanted cells remained after 37 days. After eight weeks, while explants were comprised primarily of host cells, there were also human cells attached along the periphery of BCP and embedded in osteocyte lacunae dispersed throughout the newly formed bone matrix. CONCLUSIONS This study demonstrates the safety and efficacy of BMSC/BCP combinations and provides crucial information for the implementation of BMSC therapy for bone regeneration.
Collapse
|
46
|
Allen AB, Gazit Z, Su S, Stevens HY, Guldberg RE. In vivo bioluminescent tracking of mesenchymal stem cells within large hydrogel constructs. Tissue Eng Part C Methods 2014; 20:806-16. [PMID: 24576050 DOI: 10.1089/ten.tec.2013.0587] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The use of multicomponent scaffolds for cell implantation has necessitated sophisticated techniques for tracking of cell survival in vivo. Bioluminescent imaging (BLI) has emerged as a noninvasive tool for evaluating the therapeutic potential of cell-based tissue engineering strategies. However, the ability to use BLI measurements to longitudinally assess large 3D cellular constructs in vivo and the effects of potential confounding factors are poorly understood. In this study, luciferase-expressing human mesenchymal stem cells (hMSCs) were delivered subcutaneously within agarose and RGD-functionalized alginate hydrogel vehicles to investigate the impact of construct composition and tissue formation on BLI signal. Results showed that alginate constructs exhibited twofold greater BLI counts than agarose constructs at comparable hMSC doses. However, each hydrogel type produced a linear correlation between BLI counts and live cell number, indicating that within a given material, relative differences in cell number could be accurately assessed at early time points. The survival efficiency of delivered hMSCs was highest for the lower cell doses embedded within alginate matrix. BLI signal remained predictive of live cell number through 1 week in vivo, although the strength of correlation decreased over time. Irrespective of hydrogel type or initial hMSC seeding dose, all constructs demonstrated a degree of vascularization and development of a fibrotic capsule after 1 week. Formation of tissue within and adjacent to the constructs was accompanied by an attenuation of BLI signal during the initial period of the image acquisition time-frame. In alginate constructs only, greater vessel volume led to a delayed rise in BLI signal following luciferin delivery. This study identified vascular and fibrotic tissue ingrowth as potential confounding variables for longitudinal BLI studies. Further investigation into the complexities of noninvasive BLI data acquisition from multicomponent constructs, following implantation and subsequent tissue formation, is warranted.
Collapse
Affiliation(s)
- Ashley B Allen
- 1 Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | | | | | | | | |
Collapse
|
47
|
A Comparative Evaluation between New Ternary Zirconium Alloys as Alternative Metals for Orthopedic and Dental Prosthetic Devices. Int J Artif Organs 2014; 37:149-64. [DOI: 10.5301/ijao.5000287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 11/20/2022]
Abstract
Purpose We assessed in vitro the corrosion behavior and biocompatibility of four Zr-based alloys (Zr97.5 Nb1.5 VM1.0 ; VM, valve metal: Ti, Mo, W, Ta; at%) to be used as implant materials, comparing the results with grade-2 titanium, a biocompatible metal standard. Methods Corrosion resistance was investigated by open circuit potential and electrochemical impedance spectroscopy measurements as a function of exposure time to an artificial physiological environment (Ringer's solution). Human bone marrow stromal cells were used to evaluate biocompatibility of the alloys and their influence on growth kinetics and cell osteogenic differentiation through histochemical and gene expression analyses. Results Open circuit potential values indicated that Zr-based alloys and grade-2 Ti undergo spontaneous passivation in the simulated aggressive environment. High impedance values for all samples demonstrated improved corrosion resistance of the oxide film, with the best protection characteristics displayed by Zr97.5 Nb1.5 Ta1.0. Cells seeded on all surfaces showed the same growth kinetics, although matrix mineralization and alkaline phosphatase activity were maximal on Zr97.5 Nb1.5 Mo1.0 and Zr97.5 Nb1.5 Ta1.0. Markers of ongoing proliferation, however, such as podocalyxin and CD49f, were still overexpressed on Zr97.5 Nb1.5 Mo1.0 even upon osteoinduction. No relevant effects were noted for the CD146-expressing population of bone progenitors. Nonetheless, the presence of a more differentiated cell population on Zr97.5 Nb1.5 Ta1.0 samples was inferable by comparing mineralization data and transcript levels of osteogenic markers (osteocalcin, osteopontin, bone sialoprotein, and RUNX2). Conclusions The combination of passivation, corrosion resistance and satisfactory biotolerance to bone progenitors make the Zr-based alloys promising implant materials. Among those we tested, Zr97.5 Nb1.5 Ta1.0 seems to be the most appealing.
Collapse
|
48
|
Kokemüller H, Jehn P, Spalthoff S, Essig H, Tavassol F, Schumann P, Andreae A, Nolte I, Jagodzinski M, Gellrich NC. En bloc prefabrication of vascularized bioartificial bone grafts in sheep and complete workflow for custom-made transplants. Int J Oral Maxillofac Surg 2014; 43:163-72. [DOI: 10.1016/j.ijom.2013.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/25/2013] [Accepted: 10/10/2013] [Indexed: 12/18/2022]
|
49
|
Krishnan L, Willett NJ, Guldberg RE. Vascularization strategies for bone regeneration. Ann Biomed Eng 2014; 42:432-44. [PMID: 24468975 DOI: 10.1007/s10439-014-0969-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/02/2014] [Indexed: 12/31/2022]
Abstract
The functional regeneration of thick vascularized tissues such as bone and muscle is complicated by the large volume of lost tissue, challenging biomechanical environment, and the need to reproduce the highly organized structure of both the native tissue extracellular matrix and its vascular support system. Stem cell or progenitor cell delivery approaches, for example, continue to be plagued by low viability and engraftment in part due to the initial absence of a vascular supply. Recognition of diffusion limitations in thick tissues has prompted regenerative strategies that seek to accelerate establishment of a functional vasculature. The successful design of robust regeneration strategies for these challenging clinical scenarios will rely on a thorough understanding of interactions between construct design parameters and host biological and biomechanical factors. Here, we discuss the critical role of vascularization in normal bone tissue homeostasis and repair, vascular network adaptation to the local biomechanical environment, and the future directions of revascularization approaches being developed and integrated with bone regeneration strategies.
Collapse
Affiliation(s)
- Laxminarayanan Krishnan
- Parker H. Petit Institute for Bioengineering and Bioscience, George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30332-0363, USA
| | | | | |
Collapse
|
50
|
Ma J, Both SK, Yang F, Cui FZ, Pan J, Meijer GJ, Jansen JA, van den Beucken JJJP. Concise review: cell-based strategies in bone tissue engineering and regenerative medicine. Stem Cells Transl Med 2013; 3:98-107. [PMID: 24300556 DOI: 10.5966/sctm.2013-0126] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cellular strategies play an important role in bone tissue engineering and regenerative medicine (BTE/RM). Variability in cell culture procedures (e.g., cell types, cell isolation and expansion, cell seeding methods, and preculture conditions before in vivo implantation) may influence experimental outcome. Meanwhile, outcomes from initial clinical trials are far behind those of animal studies, which is suggested to be related to insufficient nutrient and oxygen supply inside the BTE/RM constructs as some complex clinical implementations require bone regeneration in too large a quantity. Coculture strategies, in which angiogenic cells are introduced into osteogenic cell cultures, might provide a solution for improving vascularization and hence increasing bone formation for cell-based constructs. So far, preclinical studies have demonstrated that cell-based tissue-engineered constructs generally induce more bone formation compared with acellular constructs. Further, cocultures have been shown to enhance vascularization and bone formation compared with monocultures. However, translational efficacy from animal studies to clinical use requires improvement, and the role implanted cells play in clinical bone regeneration needs to be further elucidated. In view of this, the present review provides an overview of the critical procedures during in vitro and in vivo phases for cell-based strategies (both monoculture and coculture) in BTE/RM to achieve more standardized culture conditions for future studies, and hence enhance bone formation.
Collapse
Affiliation(s)
- Jinling Ma
- Department of VIP Service and Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biomaterials and Department of Oral and Maxillofacial Surgery, Radboud University Medical Center, Nijmegen, The Netherlands; State Key Laboratory of New Ceramics and Fine Processing, Department of Materials Science & Engineering, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|