Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Commun Biol ; 6(1): 1266, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38092861

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.


Subject(s)
Hydrogels , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Cell Survival , Glucose/metabolism , Starch/metabolism
2.
Adv Healthc Mater ; 12(30): e2301692, 2023 12.
Article in English | MEDLINE | ID: mdl-37655491

ABSTRACT

The reconstruction of critical-size bone defects in long bones remains a challenge for clinicians. A new osteoinductive medical device is developed here for long bone repair by combining a 3D-printed architectured cylindrical scaffold made of clinical-grade polylactic acid (PLA) with a polyelectrolyte film coating delivering the osteogenic bone morphogenetic protein 2 (BMP-2). This film-coated scaffold is used to repair a sheep metatarsal 25-mm long critical-size bone defect. In vitro and in vivo biocompatibility of the film-coated PLA material is proved according to ISO standards. Scaffold geometry is found to influence BMP-2 incorporation. Bone regeneration is followed using X-ray scans, µCT scans, and histology. It is shown that scaffold internal geometry, notably pore shape, influenced bone regeneration, which is homogenous longitudinally. Scaffolds with cubic pores of ≈870 µm and a low BMP-2 dose of ≈120 µg cm-3 induce the best bone regeneration without any adverse effects. The visual score given by clinicians during animal follow-up is found to be an easy way to predict bone regeneration. This work opens perspectives for a clinical application in personalized bone regeneration.


Subject(s)
Metatarsal Bones , Tissue Scaffolds , Animals , Sheep , Bone Regeneration , Osteogenesis , Polyesters/pharmacology , Polymers/pharmacology , Printing, Three-Dimensional , Tissue Engineering
3.
Stem Cell Rev Rep ; 19(8): 2869-2885, 2023 11.
Article in English | MEDLINE | ID: mdl-37642900

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.


Subject(s)
Adipose Tissue , Osteogenesis , Humans , Mice , Animals , Osteogenesis/genetics , Cells, Cultured , Stem Cells , Immunity, Innate
4.
Mol Ther Methods Clin Dev ; 29: 350-363, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37214314

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) is an osteogenic protein used clinically to enhance bone healing. However, it must be applied in very high doses, causing adverse side effects and increasing costs while providing only incremental benefit. Preclinical models of bone healing using gene transfer to deliver BMP-2 suggest that transgenic BMP-2 is much more osteogenic than rhBMP-2. Using a reporter mesenchymal cell line, we found transgenic human BMP-2 cDNA to be at least 100-fold more effective than rhBMP-2 in signaling. Moreover, a substantial portion of the BMP-2 produced by the transduced cells remained cell associated. Signaling by transgenic BMP-2 occurred via binding to the type I receptor, activating the associated kinase and generating phospho-smads. Signaling was partially resistant to noggin, an important extracellular inhibitor of BMP-2, possibly because nascent BMP-2 binds to its cell surface receptor during secretion and thus signals in a protected peri-cellular environment. Although the amounts of BMP-2 secreted by the transduced cells were too low to affect distant cells, transduced cells were able to induce signaling in a paracrine fashion that required close proximity of the cells, possibly cell-to-cell contact. The greater osteogenic potency of transgenic BMP-2 was confirmed with human bone marrow stromal cells.

5.
J Control Release ; 334: 188-200, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33895201

ABSTRACT

Messenger RNA (mRNA) activated matrices (RAMs) are interesting to orchestrate tissue and organ regeneration due to the in-situ and sustained production of functional proteins. However, the immunogenicity of in vitro transcribed mRNA and the paucity of proper in vivo mRNA delivery vector need to be overcome to exert the therapeutic potential of RAM. We developed a dual mRNAs system for in vitro osteogenesis by co-delivering NS1 mRNA with BMP2 mRNA to inhibit RNA sensors and enhance BMP-2 expression. Next, we evaluated a lipopolyplex (LPR) formulation platform for in vivo mRNA delivery and adapted the LPRs for RAM preparation. The LPR formulated BMP2/NS1 mRNAs were incorporated into an optimized collagen-nanohydroxyapatite scaffold and freeze-dried to prepare ready-to-use RAMs. The loaded BMP2/NS1 mRNAs lipopolyplexes maintained their spherical morphology in the RAM, thanks to the core-shell structure of LPR. The mRNAs release from RAMs lasted for 16 days resulting in an enhanced prolonged transgene expression period compared to direct cell transfection. Once subcutaneously implanted in mice, the BMP2/NS1 mRNAs LPRs containing RAMs (RAM-BMP2/NS1) induced significant new bone tissue than those without NS1 mRNA, eight weeks post implantation. Overall, our results demonstrate that the BMP2/NS1 dual mRNAs system is suitable for osteogenic engagement, and the freeze-dried RAM-BMP2/NS1 could be promising off-the-shelf products for clinical orthopedic practice.


Subject(s)
Bone Morphogenetic Protein 2 , Bone and Bones , Osteogenesis , Tissue Scaffolds , Animals , Bone Morphogenetic Protein 2/genetics , Bone Regeneration , Collagen , Durapatite , Mice , Nanoparticles , RNA, Messenger/genetics
6.
Acta Biomater ; 116: 186-200, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32911108

ABSTRACT

While human bone morphogenetic protein-2 (BMP-2) is a promising growth factor for bone regeneration, a major challenge in biomedical applications is finding an optimal carrier for its delivery at the site of injury. Because of their natural affinities for growth factors (including BMP-2) as well as their role in instructing cell function, cultured cell-derived extracellular matrices (ECM) are of special interest. We hereby hypothesized that a "bony matrix" containing mineralized, osteogenic ECM is a potential efficacious carrier of BMP-2 for promoting bone formation and, therefore, compared the efficacy of the decellularized ECM derived from osteogenic-differentiated human mesenchymal stem cells (hMSCs) to the one obtained from ECM from undifferentiated hMSCs. Our results provided evidence that both ECMs can bind BMP-2 and promote bone formation when implanted ectopically in mice. The osteoinductive potential of BMP-2, however, was greater when loaded within an osteogenic MSC-derived ECM; this outcome was correlated with higher sequestration capacity of BMP-2 over time in vivo. Interestingly, although the BMP-2 mainly bound onto the mineral crystals contained within the osteogenic MSC derived-ECM, these mineral components were not involved in the observed higher osteoinductivity, suggesting that the organic components were the critical components for the matrix efficacy as BMP-2 carrier.


Subject(s)
Mesenchymal Stem Cells , Animals , Bone Morphogenetic Protein 2 , Bone Regeneration , Cell Differentiation , Cells, Cultured , Extracellular Matrix , Mice , Osteogenesis
7.
Acta Biomater ; 108: 337-346, 2020 05.
Article in English | MEDLINE | ID: mdl-32251783

ABSTRACT

Application of messenger RNA (mRNA) for bone regeneration is a promising alternative to DNA, recombinant proteins and peptides. However, exogenous in vitro transcribed mRNA (IVT mRNA) triggers innate immune response resulting in mRNA degradation and translation inhibition. Inspired by the ability of viral immune evasion proteins to inhibit host cell responses against viral RNA, we applied non-structural protein-1 (NS1) from Influenza A virus (A/Texas/36/1991) as an IVT mRNA enhancer. We evidenced a dose-dependent blocking of RNA sensors by NS1 expression. The co-delivery of NS1 mRNA with mRNA of reporter genes significantly increased the translation efficiency. Interestingly, unlike the use of nucleosides modification, NS1-mediated mRNA translation enhancement does not dependent to cell type. Dual delivery of NS1 mRNA and BMP-2 mRNA to murine pluripotent stem cells (C3H10T1/2), promoted osteogenic differentiation evidenced by enhanced expression of osteoblastic markers (e.g. alkaline phosphatase, type I collagen, osteopontin, and osteocalcin), and extracellular mineralization. Overall, these results support the adjuvant potentiality of NS1 for mRNA-based regenerative therapies. STATEMENT OF SIGNIFICANCE: mRNA therapy has the potential to improve the efficiency of nucleic acid based regenerative medicine. Up to now, the incorporation of expensive modified nucleotides is a common way to avoid IVT mRNA-induced detrimental immunogenicity. We here introduce co-delivery of Influenza virus immune evasion protein-NS1 coding mRNA as a strategy to suppress RNA sensors for maximizing IVT mRNA expression. An increased osteogenic commitment of pluripotent stem cells was observed after BMP2 mRNA and NS1 mRNA delivery. This study revealed how applying non-modified mRNA with NS1 could be a promising alternative as a therapeutic in bone regeneration.


Subject(s)
Osteogenesis , Pluripotent Stem Cells , Animals , Bone Morphogenetic Protein 2/genetics , Cell Differentiation , Mice , RNA, Messenger/genetics , Recombinant Proteins
8.
Stem Cells ; 38(1): 22-33, 2020 01.
Article in English | MEDLINE | ID: mdl-31408238

ABSTRACT

In tissue engineering and regenerative medicine, stem cell-specifically, mesenchymal stromal/stem cells (MSCs)-therapies have fallen short of their initial promise and hype. The observed marginal, to no benefit, success in several applications has been attributed primarily to poor cell survival and engraftment at transplantation sites. MSCs have a metabolism that is flexible enough to enable them to fulfill their various cellular functions and remarkably sensitive to different cellular and environmental cues. At the transplantation sites, MSCs experience hostile environments devoid or, at the very least, severely depleted of oxygen and nutrients. The impact of this particular setting on MSC metabolism ultimately affects their survival and function. In order to develop the next generation of cell-delivery materials and methods, scientists must have a better understanding of the metabolic switches MSCs experience upon transplantation. By designing treatment strategies with cell metabolism in mind, scientists may improve survival and the overall therapeutic potential of MSCs. Here, we provide a comprehensive review of plausible metabolic switches in response to implantation and of the various strategies currently used to leverage MSC metabolism to improve stem cell-based therapeutics.


Subject(s)
Mesenchymal Stem Cells/metabolism , Regenerative Medicine/methods , Tissue Engineering/methods , Humans
9.
Int J Pharm ; 569: 118594, 2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31394184

ABSTRACT

Nucleic acid-based therapy has shown great promise in accelerating bone regeneration as well as other diseases. Nucleic acids used in gene therapy mainly are either plasmid DNA (pDNA) or RNAs. Although pDNA therapy has been extensively studied for decades with encouraging preclinical and clinical results, side effects, and low efficiency associated with nuclear trafficking are hard to bypass. Unlike pDNA, RNAs (mRNA, siRNA, miRNA) exert their function in the cytoplasm, thereby being more efficient in hard-to-transfect cells such as primary osteoblasts. RNA interference-based gene silencing represents a negative regulation which knockdown the expression of antagonists that impair osteogenesis process. In contrary, mRNA therapy for osteogenesis represents a positive regulation which delivers mRNA encoding growth factors to accelerate bone regeneration. This review presents a comprehensive summary of the mRNA and siRNA-based therapies and the targets for bone regeneration in case of bone defect and osteoporosis.


Subject(s)
Osteogenesis , RNA, Messenger/therapeutic use , RNA, Small Interfering/therapeutic use , Animals , Bone Regeneration , Bone and Bones/metabolism , Humans , Signal Transduction
10.
Tissue Eng Part A ; 25(7-8): 642-651, 2019 04.
Article in English | MEDLINE | ID: mdl-30311857

ABSTRACT

IMPACT STATEMENT: A strategy for improving the efficacy of stem cell-based bone tissue engineering (TE) constructs is to combine bone morphogenetic protein-2 (BMP-2) with multipotent stromal cells (MSC). Previous studies on the potential cooperative effect of BMP-2 with human multipotent stromal cells (hMSCs) on bone formation in vivo have, however, shown contradictory results likely due to the various and/or inappropriate BMP-2 doses. Our results provided evidence that the addition of BMP-2 at low dose only was beneficial to improve the osteogenic potential of hMSCs-containing TE constructs, whereas BMP-2 delivered at high dose overcame the advantage of combining this growth factor with hMSCs. This new knowledge will help in designing improved combination strategies for tissue regeneration with better clinical outcomes.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Tissue Engineering/methods , Bone Morphogenetic Protein 2/administration & dosage , Cells, Cultured , Humans , Osteogenesis/drug effects
11.
Mater Sci Eng C Mater Biol Appl ; 95: 343-354, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30573258

ABSTRACT

This work is devoted to the processing of bone morphogenetic protein (BMP-2) functionalized silicate substituted hydroxyapatite (SiHA) ceramic spheres. The motivation behind it is to develop injectable hydrogel/bioceramic composites for bone reconstruction applications. SiHA microspheres were shaped by spray drying and thoroughly characterized. The silicate substitution was used to provide preferred chemical sites at the ceramic surface for the covalent immobilization of BMP-2. In order to control the density and the release of the immobilized BMP-2, its grafting was performed via ethoxysilanes and polyethylene glycols. A method based on Kaiser's test was used to quantify the free amino groups of grafted organosilanes available at the ceramic surface for BMP-2 immobilization. The SiHA surface modification was investigated by means of X-ray photoelectron spectroscopy, Fourier transformed infrared spectroscopy and thermogravimetry coupled with mass spectrometry. The BMP-2 bioactivity was assessed, in vitro, by measuring the luciferase expression of a stably transfected C3H10 cell line (C3H10-BRE/Luc cells). The results provided evidence that the BMP-2 grafted onto SiHA spheres remained bioactive.


Subject(s)
Bone Morphogenetic Protein 2/chemistry , Durapatite/chemistry , Silicates/chemistry , Animals , Cell Line, Tumor , Mass Spectrometry , Mice , Photoelectron Spectroscopy , Polyethylene Glycols/chemistry , Tissue Scaffolds/chemistry
12.
J Tissue Eng Regen Med ; 12(3): e1511-e1524, 2018 03.
Article in English | MEDLINE | ID: mdl-28875591

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.


Subject(s)
Adipogenesis , Bone and Bones/physiology , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Adipogenesis/drug effects , Animals , Biomarkers/metabolism , Bone and Bones/drug effects , Cell Adhesion/drug effects , Cell Lineage/drug effects , Cell Survival/drug effects , Chemotactic Factors/pharmacology , Coculture Techniques , Female , Humans , Ischemia/pathology , Mesenchymal Stem Cells/ultrastructure , Mice, Nude , Neovascularization, Physiologic/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects
13.
Stem Cells ; 36(3): 363-376, 2018 03.
Article in English | MEDLINE | ID: mdl-29266629

ABSTRACT

Mesenchymal stem cells (MSCs) hold considerable promise in tissue engineering (TE). However, their poor survival when exogenously administered limits their therapeutic potential. Previous studies from our group demonstrated that lack of glucose (glc) (but not of oxygen) is fatal to human MSCs because it serves as a pro-survival and pro-angiogenic molecule for human MSCs (hMSCs) upon transplantation. However, which energy-providing pathways MSCs use to metabolize glc upon transplantation? Are there alternative energetic nutrients to replace glc? And most importantly, do hMSCs possess significant intracellular glc reserves for ensuring their survival upon transplantation? These remain open questions at the forefront of TE based-therapies. In this study, we established for the first time that the in vivo environment experienced by hMSCs is best reflected by near-anoxia (0.1% O2 ) rather than hypoxia (1%-5% O2 ) in vitro. Under these near-anoxia conditions, hMSCs rely almost exclusively on glc through anerobic glycolysis for ATP production and are unable to use either exogenous glutamine, serine, or pyruvate as energy substrates. Most importantly, hMSCs are unable to adapt their metabolism to the lack of exogenous glc, possess a very limited internal stock of glc and virtually no ATP reserves. This lack of downregulation of energy turnover as a function of exogenous glc level results in a rapid depletion of hMSC energy reserves that explains their poor survival rate. These new insights prompt for the development of glc-releasing scaffolds to overcome this roadblock plaguing the field of TE based-therapies. Stem Cells 2018;36:363-376.


Subject(s)
Cell Survival/physiology , Glucose/metabolism , Glycolysis/physiology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Cell Differentiation/physiology , Cell Hypoxia/physiology , Glutamine/metabolism , Humans , Oxygen/metabolism , Tissue Engineering
14.
J Biol Chem ; 292(37): 15352-15368, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28747434

ABSTRACT

Bone morphogenetic proteins (BMPs) regulate diverse cellular responses during embryogenesis and in adulthood including cell differentiation, proliferation, and death in various tissues. In the adult pituitary, BMPs participate in the control of hormone secretion and cell proliferation, suggesting a potential endocrine/paracrine role for BMPs, but some of the mechanisms are unclear. Here, using a bioactivity test based on embryonic cells (C3H10T1/2) transfected with a BMP-responsive element, we sought to determine whether pituitary cells secrete BMPs or BMP antagonists. Interestingly, we found that pituitary-conditioned medium contains a factor that inhibits action of BMP-2 and -4. Combining surface plasmon resonance and high-resolution mass spectrometry helped pinpoint this factor as thrombospondin-1 (TSP-1). Surface plasmon resonance and co-immunoprecipitation confirmed that recombinant human TSP-1 can bind BMP-2 and -4 and antagonize their effects on C3H10T1/2 cells. Moreover, TSP-1 inhibited the action of serum BMPs. We also report that the von Willebrand type C domain of TSP-1 is likely responsible for this BMP-2/4-binding activity, an assertion based on sequence similarity that TSP-1 shares with the von Willebrand type C domain of Crossveinless 2 (CV-2), a BMP antagonist and member of the chordin family. In summary, we identified for the first time TSP-1 as a BMP-2/-4 antagonist and presented a structural basis for the physical interaction between TSP-1 and BMP-4. We propose that TSP-1 could regulate bioavailability of BMPs, either produced locally or reaching the pituitary via blood circulation. In conclusion, our findings provide new insights into the involvement of TSP-1 in the BMP-2/-4 mechanisms of action.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 4/antagonists & inhibitors , Models, Molecular , Pituitary Gland/metabolism , Response Elements , Thrombospondin 1/metabolism , Animals , Animals, Inbred Strains , Bone Morphogenetic Protein 2/blood , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/blood , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Line , Cells, Cultured , Computational Biology , Female , Genes, Reporter , Humans , Mice , Pituitary Gland/cytology , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sheep, Domestic , Thrombospondin 1/chemistry , Thrombospondin 1/isolation & purification
15.
J Orthop Res ; 35(12): 2637-2645, 2017 12.
Article in English | MEDLINE | ID: mdl-28401593

ABSTRACT

Tissue-engineered constructs (TECs) combining resorbable calcium-based scaffolds and mesenchymal stem cells (MSCs) have the capability to regenerate large bone defects. Inconsistent results have, however, been observed, with a lack of osteoinductivity as a possible cause of failure. This study aimed to evaluate the impact of the addition of low-dose bone morphogenetic protein-2 (BMP-2) to MSC-coral-TECs on the healing of clinically relevant segmental bone defects in sheep. Coral granules were either seeded with autologous MSCs (bone marrow-derived) or loaded with BMP-2. A 25-mm-long metatarsal bone defect was created and stabilized with a plate in 18 sheep. Defects were filled with one of the following TECs: (i) BMP (n = 5); (ii) MSC (n = 7); or (iii) MSC-BMP (n = 6). Radiographic follow-up was performed until animal sacrifice at 4 months. Bone formation and scaffold resorption were assessed by micro-CT and histological analysis. Bone union with nearly complete scaffold resorption was observed in 1/5, 2/7, and 3/6 animals, when BMP-, MSC-, and MSC-BMP-TECs were implanted, respectively. The amount of newly formed bone was not statistically different between groups: 1074 mm3 [970-2478 mm3 ], 1155 mm3 [970-2595 mm3 ], and 2343 mm3 [931-3276 mm3 ] for BMP-, MSC-, and MSC-BMP-TECs, respectively. Increased scaffold resorption rate using BMP-TECs was the only potential side effect observed. In conclusion, although the dual delivery of MSCs and BMP-2 onto a coral scaffold further increased bone formation and bone union when compared to single treatment, results were non-significant. Only 50% of the defects healed, demonstrating the need for further refinement of this strategy before clinical use. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2637-2645, 2017.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Bone Regeneration/drug effects , Mesenchymal Stem Cell Transplantation , Tissue Scaffolds , Animals , Anthozoa , Drug Evaluation, Preclinical , Female , Guided Tissue Regeneration , Metatarsal Bones , Sheep
16.
Stem Cells ; 35(1): 181-196, 2017 01.
Article in English | MEDLINE | ID: mdl-27578059

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.


Subject(s)
Cell Cycle , Ischemia/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Metabolome , Adenosine Triphosphate/metabolism , Autophagy , Cell Cycle Checkpoints , Cell Survival , Cells, Cultured , Culture Media, Serum-Free , Humans , Mesenchymal Stem Cell Transplantation , Reperfusion , Stress, Physiological
17.
Tissue Eng Part A ; 22(5-6): 534-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26896389

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.


Subject(s)
Choristoma/pathology , Femur/pathology , Mesenchymal Stem Cells/cytology , Osteogenesis , Animals , Bone Resorption/pathology , Cell Proliferation , Cell Survival , Cell Tracking , Densitometry , Humans , Implants, Experimental , Luciferases/metabolism , Male , Mice, Nude , Phenotype , Tissue Scaffolds/chemistry
18.
J Tissue Eng Regen Med ; 10(3): E177-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-23784976

ABSTRACT

Tissue-engineered constructs combining bone marrow mesenchymal stem cells with biodegradable osteoconductive scaffolds are very promising for repairing large segmental bone defects. Synchronizing and controlling the balance between scaffold-material resorption and new bone tissue formation are crucial aspects for the success of bone tissue engineering. The purpose of the present study was to determine, and compare, the osteogenic potential of ceramic scaffolds with different resorbability. Four clinically relevant granular biomaterial scaffolds (specifically, Porites coral, Acropora coral, beta-tricalcium phosphate and banked bone) with or without autologous bone marrow stromal cells were implanted in the ectopic, subcutaneous-pouch sheep model. Scaffold material resorption and new bone formation were assessed eight weeks after implantation. New bone formation was only detected when the biomaterial constructs tested contained MSCs. New bone formation was higher in the Porites coral and Acropora coral than in either the beta-tricalcium phosphate or the banked bone constructs; furthermore, there was a direct correlation between scaffold resorption and bone formation. The results of the present study provide evidence that, among the biomaterials tested, coral scaffolds containing MSCs promoted the best new bone formation in the present study.


Subject(s)
Ceramics/pharmacology , Osteogenesis/drug effects , Animals , Anthozoa , Bone Resorption/diagnostic imaging , Bone Resorption/pathology , Disease Models, Animal , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Sheep , Tissue Scaffolds/chemistry
19.
Stem Cells Transl Med ; 4(7): 809-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25979862

ABSTRACT

UNLABELLED: : Mesenchymal stem cells (MSCs) have captured the attention and research endeavors of the scientific world because of their differentiation potential. However, there is accumulating evidence suggesting that the beneficial effects of MSCs are predominantly due to the multitude of bioactive mediators secreted by these cells. Because the paracrine potential of MSCs is closely related to their microenvironment, the present study investigated and characterized select aspects of the human MSC (hMSC) secretome and assessed its in vitro and in vivo bioactivity as a function of oxygen tension, specifically near anoxia (0.1% O2) and hypoxia (5% O2), conditions that reflect the environment to which MSCs are exposed during MSC-based therapies in vivo. In contrast to supernatant conditioned media (CM) obtained from hMSCs cultured at either 5% or 21% of O2, CM from hMSCs cultured under near anoxia exhibited significantly (p < .05) enhanced chemotactic and proangiogenic properties and a significant (p < .05) decrease in the inflammatory mediator content. An analysis of the hMSC secretome revealed a specific profile under near anoxia: hMSCs increase their paracrine expression of the angiogenic mediators vascular endothelial growth factor (VEGF)-A, VEGF-C, interleukin-8, RANTES, and monocyte chemoattractant protein 1 but significantly decrease expression of several inflammatory/immunomodulatory mediators. These findings provide new evidence that elucidates aspects of great importance for the use of MSCs in regenerative medicine and could contribute to improving the efficacy of such therapies. SIGNIFICANCE: The present study investigated and characterized select aspects of the human mesenchymal stem cell (hMSC) secretome and assessed its in vitro and in vivo biological bioactivity as a function of oxygen tension, specifically near anoxia (0.1% O2) and hypoxia (5% O2), conditions that reflect the environment to which MSCs are exposed during MSC-based therapies in vivo. The present study provided the first evidence of a shift of the hMSC cytokine signature induced by oxygen tension, particularly near anoxia (0.1% O2). Conditioned media obtained from hMSCs cultured under near anoxia exhibited significantly enhanced chemotactic and proangiogenic properties and a significant decrease in the inflammatory mediator content. These findings provide new evidence that elucidates aspects of great importance for the use of MSCs in regenerative medicine, could contribute to improving the efficacy of such therapies, and most importantly highlighted the interest in using conditioned media in therapeutic modalities.

20.
Tissue Eng Part A ; 20(13-14): 1827-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24447025

ABSTRACT

The present study aimed at elucidating the effect of local pH in the extracellular microenvironment of tissue-engineered (TE) constructs on bone cell functions pertinent to new tissue formation. To this aim, we evaluated the osteogenicity process associated with bone constructs prepared from human Bone marrow-derived mesenchymal stem cells (hBMSC) combined with 45S5 bioactive glass (BG), a material that induces alkalinization of the external medium. The pH measured in cell-containing BG constructs was around 8.0, that is, 0.5 U more alkaline than that in two other cell-containing materials (hydroxyapatite/tricalcium phosphate [HA/TCP] and coral) constructs tested. When implanted ectopically in mice, there was no de novo bone tissue in the BG cell-containing constructs, in contrast to results obtained with either HA/TCP or coral ceramics, which consistently promoted the formation of ectopic bone. In addition, the implanted 50:50 composites of both HA/TCP:BG and coral:BG constructs, which displayed a pH of around 7.8, promoted 20-30-fold less amount of bone tissue. Interestingly, hBMSC viability in BG constructs was not affected compared with the other two types of material constructs tested both in vitro and in vivo. Osteogenic differentiation (specifically, the alkaline phosphatase [ALP] activity and gene expression of RUNX2, ALP, and BSP) was not affected when hBMSC were maintained in moderate alkaline pH (≤7.90) external milieu in vitro, but was dramatically inhibited at higher pH values. The formation of mineralized nodules in the extracellular matrix of hBMSC was fully inhibited at alkaline (>7.54) pH values. Most importantly, there is a pH range (specifically, 7.9-8.27) at which hBMSC proliferation was not affected, but the osteogenic differentiation of these cells was inhibited. Altogether, these findings provided evidence that excessive alkalinization in the microenvironment of TE constructs (resulting, for example, from material degradation) affects adversely the osteogenic differentiation of osteoprogenitor cells.


Subject(s)
Cellular Microenvironment , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adolescent , Adult , Animals , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cellular Microenvironment/drug effects , Culture Media , Female , Humans , Hydrogen-Ion Concentration/drug effects , Implants, Experimental , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Nude , Microscopy, Electron, Scanning , Middle Aged , Osteogenesis/drug effects , Subcutaneous Tissue/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...