Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(19)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33008121

ABSTRACT

Most bones of the human body form and heal through endochondral ossification, whereby hypertrophic cartilage (HyC) is formed and subsequently remodeled into bone. We previously demonstrated that HyC can be engineered from human mesenchymal stromal cells (hMSC), and subsequently devitalized by apoptosis induction. The resulting extracellular matrix (ECM) tissue retained osteoinductive properties, leading to ectopic bone formation. In this study, we aimed at engineering and devitalizing upscaled quantities of HyC ECM within a perfusion bioreactor, followed by in vivo assessment in an orthotopic bone repair model. We hypothesized that the devitalized HyC ECM would outperform a clinical product currently used for bone reconstructive surgery. Human MSC were genetically engineered with a gene cassette enabling apoptosis induction upon addition of an adjuvant. Engineered hMSC were seeded, differentiated, and devitalized within a perfusion bioreactor. The resulting HyC ECM was subsequently implanted in a 10-mm rabbit calvarial defect model, with processed human bone (Maxgraft®) as control. Human MSC cultured in the perfusion bioreactor generated a homogenous HyC ECM and were efficiently induced towards apoptosis. Following six weeks of in vivo implantation, microcomputed tomography and histological analyses of the defects revealed an increased bone formation in the defects filled with HyC ECM as compared to Maxgraft®. This work demonstrates the suitability of engineered devitalized HyC ECM as a bone substitute material, with a performance superior to a state-of-the-art commercial graft. Streamlined generation of the devitalized tissue transplant within a perfusion bioreactor is relevant towards standardized and automated manufacturing of a clinical product.


Subject(s)
Cartilage/growth & development , Cell Differentiation/genetics , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Skull/growth & development , Animals , Apoptosis/genetics , Bone Remodeling/genetics , Bone Substitutes/therapeutic use , Cartilage/metabolism , Cartilage/transplantation , Extracellular Matrix/genetics , Humans , Mesenchymal Stem Cell Transplantation , Osteogenesis/genetics , Rabbits , Skull/physiopathology , Skull/surgery , Tissue Engineering/methods , Tissue Scaffolds , Wound Healing/genetics
2.
Adv Biosyst ; 3(4): e1800300, 2019 04.
Article in English | MEDLINE | ID: mdl-32627426

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death. Conventional chemotherapeutic regimens have limited success rates, and a major challenge for the development of novel therapies is the lack of adequate in vitro models. Nonmalignant mesenchymal and immune cells of the tumor microenvironment (TME) are known to critically affect CRC progression and drug responsiveness. However, tumor drug sensitivity is still evaluated on systems, such as cell monolayers, spheroids, or tumor xenografts, which typically neglect the original TME. Here, it is investigated whether a bioreactor-based 3D culture system can preserve the main TME cellular components in primary CRC samples. Freshly excised CRC fragments are inserted between two collagen scaffolds in a "sandwich-like" format and cultured under static or perfused conditions up to 3 d. Perfused cultures maintain tumor tissue architecture and densities of proliferating tumor cells to significantly higher extents than static cultures. Stromal and immune cells are also preserved and fully viable, as indicated by their responsiveness to microenvironmental stimuli. Importantly, perfusion-based cultures prove suitable for testing the sensitivity of primary tumor cells to chemotherapies currently in use for CRC. Perfusion-based culture of primary CRC specimens recapitulates TME key features and may allow assessment of tumor drug response in a patient-specific context.


Subject(s)
Bioreactors , Cell Culture Techniques , Colorectal Neoplasms/metabolism , Tumor Microenvironment/physiology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Collagen , Colorectal Neoplasms/pathology , Equipment Design , Humans , Perfusion , Spheroids, Cellular/physiology , Tissue Scaffolds/chemistry
3.
Tissue Eng Part C Methods ; 24(7): 391-398, 2018 07.
Article in English | MEDLINE | ID: mdl-29897015

ABSTRACT

Triggering receptor expressed on myeloid cells-2 (TREM-2), a transmembrane receptor expressed by macrophages, microglia, and osteoclasts (OCs), plays a protective role in late-onset Alzheimer Disease (AD). To validate TREM-2 as a therapeutic target in AD, its potential secondary parallel effect on bone homeostasis should be clarified. However, animal models and monolayer cultures of human cells were shown poorly predictive of TREM-2 function in human. Therefore, this study aimed to engineer a tridimensional in vitro model using human progenitors differentiated into osteoblasts and OCs, recapitulating physiological bone homeostasis. Human bone marrow-derived mesenchymal cells were seeded and cultured under perfusion inside a collagen type I scaffold for 3 weeks, generating osteoblasts and mineralized matrix. Human peripheral blood-derived CD14+ monocytes were subsequently seeded through the generated tissue, thanks to perfusion flow, and further cultured for up to 3 weeks with an inductive medium, generating mature OCs. This culture system supported collagenous matrix deposition and resorption, allowing for the investigation of kinetic of soluble TREM-2 over the coculture time. Agonistic activation of TREM-2 in this model had no effect on OC activity or on mineralized matrix turnover. In conclusion, the engineered culture system represents a tridimensional, in vitro human bone model for drug testing and suggested no effect of TREM-2 agonist on bone resorption.


Subject(s)
Bone and Bones/cytology , Homeostasis , Membrane Glycoproteins/metabolism , Models, Biological , Osteoclasts/cytology , Receptors, Immunologic/metabolism , Tissue Engineering , Bone and Bones/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Osteoclasts/metabolism
4.
Tissue Eng Part A ; 23(13-14): 708-715, 2017 07.
Article in English | MEDLINE | ID: mdl-28338424

ABSTRACT

Engineered hypertrophic cartilage (HC) represents an attractive bone substitute material, capable to induce bone formation by endochondral ossification. Since bone formation by HC depends on factors released from the extracellular matrix, in this study, we hypothesized that HC seeding with monocytes committed to osteoclastogenesis could enhance its remodeling, improve chemotaxis of skeletal and vascular cells, and consequently enhance bone formation. This would be particularly relevant for devitalized HC, which currently exhibits only limited osteoinductivity. Living or devitalized HC engineered from human bone marrow-derived mesenchymal stromal cells (MSCs) was seeded or not with human monocytes in the presence of macrophage colony-stimulating factor and RANK-ligand, cultured for up to 15 days, or implanted ectopically in nude mice. Monocytes seeded on devitalized, but not living, HC induced its in vitro resorption, resulting in 30-fold higher release and 2.7-fold lower content of glycosaminoglycans compared with unseeded samples. In vitro, supernatants from monocyte-seeded devitalized HC attracted more monocytes compared with unseeded samples, but did not enhance chemotaxis of MSCs or human umbilical vein endothelial cells. In vivo, however, neither remodeling nor invasion by osteoclasts, endothelial cells, and mouse MSCs were significantly affected by the seeding with monocytes. Finally, in vitro priming of living or devitalized HC by monocytes did not enhance their bone-forming capacity. Further investigations should test the proposed approach on HC engineered to prevent rapid degradation and support osteoclastogenesis, or identify alternative strategies to enhance engineered HC remodeling and bone-forming capacity.


Subject(s)
Cartilage/chemistry , Monocytes/metabolism , Osteogenesis , Adult , Animals , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Monocytes/cytology , Monocytes/transplantation
5.
Methods Mol Biol ; 1416: 413-24, 2016.
Article in English | MEDLINE | ID: mdl-27236686

ABSTRACT

Bone development, growth, and repair predominantly occur through the process of endochondral ossification, characterized by remodelling of cartilaginous templates. The same route efficiently supports engineering of bone marrow as a niche for hematopoietic stem cells (HSC). Here we describe a combined in vitro/in vivo system based on bone marrow-derived Mesenchymal Stem/Stromal Cells (MSC) that duplicates the hallmark cellular and molecular events of endochondral ossification during development. The model requires MSC culture with instructive molecules to generate hypertrophic cartilage tissues. The resulting constructs complete the endochondral route upon in vivo implantation, in the timeframe of up to 12 weeks. The described protocol is clearly distinct from the direct ossification approach typically used to drive MSC towards osteogenesis. Recapitulation of endochondral ossification allows modelling of stromal-HSC interactions in physiology and pathology and allows engineering processes underlying bone regeneration.


Subject(s)
Adult Stem Cells/cytology , Cartilage/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Adult , Animals , Bone Transplantation , Cell Culture Techniques/methods , Cells, Cultured , Diastasis, Bone , Humans , Mice , Tissue Scaffolds
6.
Biomaterials ; 62: 138-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26051518

ABSTRACT

Anticancer compound screening on 2D cell cultures poorly predicts "in vivo" performance, while conventional 3D culture systems are usually characterized by limited cell proliferation, failing to produce tissue-like-structures (TLS) suitable for drug testing. We addressed engineering of TLS by culturing cancer cells in porous scaffolds under perfusion flow. Colorectal cancer (CRC) HT-29 cells were cultured in 2D, on collagen sponges in static conditions or in perfused bioreactors, or injected subcutaneously in immunodeficient mice. Perfused 3D (p3D) cultures resulted in significantly higher (p < 0.0001) cell proliferation than static 3D (s3D) cultures and yielded more homogeneous TLS, with morphology and phenotypes similar to xenografts. Transcriptome analysis revealed a high correlation between xenografts and p3D cultures, particularly for gene clusters regulating apoptotic processes and response to hypoxia. Treatment with 5-Fluorouracil (5-FU), a frequently used but often clinically ineffective chemotherapy drug, induced apoptosis, down-regulation of anti-apoptotic genes (BCL-2, TRAF1, and c-FLIP) and decreased cell numbers in 2D, but only "nucleolar stress" in p3D and xenografts. Conversely, BCL-2 inhibitor ABT-199 induced cytotoxic effects in p3D but not in 2D cultures. Our findings advocate the importance of perfusion flow in 3D cultures of tumor cells to efficiently mimic functional features observed "in vivo" and to test anticancer compounds.


Subject(s)
Bioreactors , Drug Resistance, Neoplasm , Fluorouracil/therapeutic use , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Tissue Engineering/instrumentation , Antimetabolites, Antineoplastic/therapeutic use , Batch Cell Culture Techniques/instrumentation , Biomimetics/methods , Cell Proliferation/drug effects , Equipment Design , Equipment Failure Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Phenotype
7.
Virulence ; 6(3): 265-73, 2015.
Article in English | MEDLINE | ID: mdl-25587671

ABSTRACT

Periodontal infection involves a complex interplay between oral biofilms, gingival tissues and cells of the immune system in a dynamic microenvironment. A humanized in vitro model that reduces the need for experimental animal models, while recapitulating key biological events in a periodontal pocket, would constitute a technical advancement in the study of periodontal disease. The aim of this study was to use a dynamic perfusion bioreactor in order to develop a gingival epithelial-fibroblast-monocyte organotypic co-culture on collagen sponges. An 11 species subgingival biofilm was used to challenge the generated tissue in the bioreactor for a period of 24 h. The histological and scanning electron microscopy analysis displayed an epithelial-like layer on the surface of the collagen sponge, supported by the underlying ingrowth of gingival fibroblasts, while monocytic cells were also found within the sponge mass. Bacterial quantification of the biofilm showed that in the presence of the organotypic tissue, the growth of selected biofilm species, especially Campylobacter rectus, Actinomyces oris, Streptococcus anginosus, Veillonella dispar, and Porphyromonas gingivalis, was suppressed, indicating a potential antimicrobial effect by the tissue. Multiplex immunoassay analysis of cytokine secretion showed that interleukin (IL)-1 ß, IL-2, IL-4, and tumor necrosis factor (TNF)-α levels in cell culture supernatants were significantly up-regulated in presence of the biofilm, indicating a positive inflammatory response of the organotypic tissue to the biofilm challenge. In conclusion, this novel host-biofilm interaction organotypic model might resemble the periodontal pocket and have an important impact on the study of periodontal infections, by minimizing the need for the use of experimental animal models.


Subject(s)
Gingiva/microbiology , Periodontal Pocket/microbiology , Biofilms/growth & development , Bioreactors , Cell Line , Coculture Techniques , Collagen , Fibrocartilage , Humans , Interleukins/metabolism , Organ Culture Techniques , Perfusion , Porphyromonas gingivalis/growth & development , Tumor Necrosis Factor-alpha/metabolism
8.
Tissue Eng Part C Methods ; 21(4): 394-403, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25226356

ABSTRACT

Enzymatic isolation of chondrocytes from a cartilage biopsy is the first step to establish in vitro models of chondrogenesis or to generate cell-based grafts for cartilage repair. Such process is based on manually operated procedures and typically results in yields lower than 20% of the total available cells. In this study, we hypothesized that, as compared to conventionally used protocols, the enzymatic digestion of human articular cartilage in the presence of ascorbic acid 2-phosphate (AscA2P) or of sodium chloride (NaCl), in combination with the use of a perfusion bioreactor system, leads to a higher and more reproducible yield of cell populations with high proliferation and chondrogenic capacity. The addition of AscA2P within the enzymatic digestion medium did not significantly increase the cell yield, but resulted in a significant decrease of the intradonor variability in cell yield (-17.8% ± 10.7%, p = 0.0247) and in a significant increase of the proliferation rate of the isolated chondrocytes (+19.0% ± 1.4%, p < 0.05) with respect to the control group. The addition of NaCl during cartilage digestion did not modulate cell yield. When the cartilage digestion was further performed under direct perfusion flow, beneficial synergistic effects were achieved, with an overall increase of 34.7% ± 6.8% (p < 0.001) in the cell yield and an average decrease of 57.8% ± 11.2% (p < 0.01) in the coefficient of variation with respect to the control group. Importantly, by implementing this strategy it was possible to retrieve clonal subpopulations more efficiently capable of undergoing chondrogenesis, both in vitro and in vivo. Our findings bear relevance for the preparation of human chondrocytes for laboratory investigations, and in the perspective of efficient and streamlined manufacturing of cell/tissue grafts for articular cartilage repair.


Subject(s)
Bioreactors , Cartilage, Articular/chemistry , Cartilage, Articular/cytology , Chondrocytes/chemistry , Chondrocytes/cytology , Aged , Aged, 80 and over , Ascorbic Acid/chemistry , Cell Separation/methods , Female , Humans , Male , Middle Aged , Sodium Chloride/chemistry
9.
Bone ; 70: 66-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25260931

ABSTRACT

Despite the significant progress in the field of bone tissue engineering, cell-based products have not yet reached the stage of clinical adoption. This is due to the uncertain advantages from the standard-of-care, combined with challenging cost-and regulatory-related issues. Novel therapeutic approaches could be based on exploitation of the intrinsic regenerative capacity of bone tissue, provided the development of a deeper understanding of its healing mechanisms. While it is well-established that endogenous progenitors can be activated toward bone formation by overdoses of single morphogens, the challenge to stimulate the healing processes by coordinated and controlled stimulation of specific cell populations remains open. Here, we review the recent approaches to generate osteoinductive materials based on the use of decellularized extracellular matrices (ECM) as reservoirs of multiple factors presented at physiological doses and through the appropriate ligands. We then propose the generation of customized engineered and decellularized ECM (i) as a tool to better understand the processes of bone regeneration and (ii) as safe and effective "off-the-shelf" bone grafts for clinical use. This article is part of a Special Issue entitled Stem Cells and Bone.


Subject(s)
Bone Regeneration/physiology , Extracellular Matrix/metabolism , Tissue Engineering/methods , Animals , Humans , Reference Standards , Wound Healing
10.
PLoS One ; 9(7): e102359, 2014.
Article in English | MEDLINE | ID: mdl-25020062

ABSTRACT

Mesenchymal stromal/stem cell (MSC) expansion in conventional monolayer culture on plastic dishes (2D) leads to progressive loss of functionality and thus challenges fundamental studies on the physiology of skeletal progenitors, as well as translational applications for cellular therapy and molecular medicine. Here we demonstrate that 2D MSC expansion can be entirely bypassed by culturing freshly isolated bone marrow nucleated cells within 3D porous scaffolds in a perfusion-based bioreactor system. The 3D-perfusion system generated a stromal tissue that could be enzymatically treated to yield CD45- MSC. As compared to 2D-expanded MSC (control), those derived from 3D-perfusion culture after the same time (3 weeks) or a similar extent of proliferation (7-8 doublings) better maintained their progenitor properties, as assessed by a 4.3-fold higher clonogenicity and the superior differentiation capacity towards all typical mesenchymal lineages. Transcriptomic analysis of MSC from 5 donors validated the robustness of the process and indicated a reduced inter-donor variability and a significant upregulation of multipotency-related gene clusters following 3D-perfusion--as compared to 2D-expansion. Interestingly, the differences in functionality and transcriptomics between MSC expanded in 2D or under 3D-perfusion were only partially captured by cytofluorimetric analysis using conventional surface markers. The described system offers a multidisciplinary approach to study how factors of a 3D engineered niche regulate MSC function and, by streamlining conventional labor-intensive processes, is prone to automation and scalability within closed bioreactor systems.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Perfusion/methods , Bioreactors , Cell Culture Techniques/instrumentation , Cell Proliferation , Cell Separation , Humans , Oligonucleotide Array Sequence Analysis , Perfusion/instrumentation , Phenotype
11.
Adv Drug Deliv Rev ; 79-80: 145-54, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24819215

ABSTRACT

Interaction between cancer cells and immune system critically affects development, progression and treatment of human malignancies. Experimental animal models and conventional "in vitro" studies have provided a wealth of information on this interaction, currently used to develop immune-mediated therapies. Studies utilizing three-dimensional culture technologies have emphasized that tumor architecture dramatically influences cancer cell-immune system interaction by steering cytokine production and regulating differentiation patterns of myeloid cells, and decreasing the sensitivity of tumor cells to lymphocyte effector functions. Hypoxia and increased production of lactic acid by tumor cells cultured in 3D architectures appear to be mechanistically involved. 3D culture systems could be further developed to (i) include additional cell partners potentially influencing cancer cell-immune system interaction, (ii) enable improved control of hypoxia, and (iii) allow the use of freshly derived clinical cancer specimens. Such advanced models will represent new tools for cancer immunobiology studies and for pre-clinical assessment of innovative treatments.


Subject(s)
Models, Biological , Neoplasms/immunology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Hypoxia/immunology , Cytokines/immunology , Disease Progression , Humans , In Vitro Techniques , Lactic Acid/metabolism , Neoplasms/pathology , Neoplasms/therapy
12.
Proc Natl Acad Sci U S A ; 110(10): 3997-4002, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23401508

ABSTRACT

Embryonic development, lengthening, and repair of most bones proceed by endochondral ossification, namely through formation of a cartilage intermediate. It was previously demonstrated that adult human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) can execute an endochondral program and ectopically generate mature bone. Here we hypothesized that hMSCs pushed through endochondral ossification can engineer a scaled-up ossicle with features of a "bone organ," including physiologically remodeled bone, mature vasculature, and a fully functional hematopoietic compartment. Engineered hypertrophic cartilage required IL-1ß to be efficiently remodeled into bone and bone marrow upon subcutaneous implantation. This model allowed distinguishing, by analogy with bone development and repair, an outer, cortical-like perichondral bone, generated mainly by host cells and laid over a premineralized area, and an inner, trabecular-like, endochondral bone, generated mainly by the human cells and formed over the cartilaginous template. Hypertrophic cartilage remodeling was paralleled by ingrowth of blood vessels, displaying sinusoid-like structures and stabilized by pericytic cells. Marrow cavities of the ossicles contained phenotypically defined hematopoietic stem cells and progenitor cells at similar frequencies as native bones, and marrow from ossicles reconstituted multilineage long-term hematopoiesis in lethally irradiated mice. This study, by invoking a "developmental engineering" paradigm, reports the generation by appropriately instructed hMSC of an ectopic "bone organ" with a size, structure, and functionality comparable to native bones. The work thus provides a model useful for fundamental and translational studies of bone morphogenesis and regeneration, as well as for the controlled manipulation of hematopoietic stem cell niches in physiology and pathology.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Tissue Engineering/methods , Adult , Animals , Bone Marrow/physiology , Bone Marrow Transplantation , Cartilage/transplantation , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Interleukin-1beta/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Biological , Neovascularization, Physiologic , Osteogenesis/drug effects , Regenerative Medicine/methods , Stem Cell Niche/physiology , Tissue Scaffolds , Transplantation, Heterologous
13.
Eur Cell Mater ; 24: 224-36, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-23007908

ABSTRACT

Inflammatory cytokines present in the milieu of the fracture site are important modulators of bone healing. Here we investigated the effects of interleukin-1ß (IL-1ß) on the main events of endochondral bone formation by human bone marrow mesenchymal stromal cells (BM-MSC), namely cell proliferation, differentiation and maturation/remodelling of the resulting hypertrophic cartilage. Low doses of IL-1ß (50 pg/mL) enhanced colony-forming units-fibroblastic (CFU-f) and -osteoblastic (CFU-o) number (up to 1.5-fold) and size (1.2-fold) in the absence of further supplements and glycosaminoglycan accumulation (1.4-fold) upon BM-MSC chondrogenic induction. In osteogenically cultured BM-MSC, IL-1ß enhanced calcium deposition (62.2-fold) and BMP-2 mRNA expression by differential activation of NF-κB and ERK signalling. IL-1ß-treatment of BM-MSC generated cartilage resulted in higher production of MMP-13 (14.0-fold) in vitro, mirrored by an increased accumulation of the cryptic cleaved fragment of aggrecan, and more efficient cartilage remodelling/resorption after 5 weeks in vivo (i.e., more TRAP positive cells and bone marrow, less cartilaginous areas), resulting in the formation of mature bone and bone marrow after 12 weeks. In conclusion, IL-1ß finely modulates early and late events of the endochondral bone formation by BM-MSC. Controlling the inflammatory environment could enhance the success of therapeutic approaches for the treatment of fractures by resident MSC and as well as improve the engineering of implantable tissues.


Subject(s)
Chondrogenesis , Interleukin-1beta/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis , Adult , Animals , Bone Morphogenetic Protein 2/genetics , Calcium/metabolism , Cartilage/growth & development , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Glycosaminoglycans/metabolism , Humans , Male , Matrix Metalloproteinase 13/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Middle Aged , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , RNA, Messenger/biosynthesis
14.
Stem Cells ; 30(7): 1455-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22495904

ABSTRACT

Bone marrow (BM) mesenchymal stem/stromal cells (MSC) are a heterogeneous population of multipotent progenitors currently under investigation for a variety of applications in regenerative medicine. While self-renewal of stem cells in different tissues has been demonstrated to be regulated by specialized microenvironments called niches, it is still unclear whether a self-renewing niche also exists for MSC. Here, we show that primary human BM cultures contain a population of intrinsically non-adherent mesenchymal progenitors (NAMP) with features of more primitive progenitors than the initially adhering colony-forming units-fibroblast (CFU-f). In fact, NAMP could generate an adherent progeny: (a) enriched with early mesenchymal populations (CD146+, SSEA-1+, and SSEA-4+); (b) with significantly greater proliferation and multilineage differentiation potential in vitro; and (c) capable of threefold greater bone formation in vivo than the corresponding CFU-f. Upon serial replating, NAMP were able to regenerate and expand in suspension as non-adherent clonogenic progenitors, while also giving rise to an adherent progeny. This took place at the cost of a gradual loss of proliferative potential, shown by a reduction in colony size, which could be completely prevented when NAMP were expanded on the initially adhering BM fraction. Mechanistically, we found that NAMP crucially depend on fibroblast growth factor (FGF)-2 signaling through FGFR2c for their survival and expansion. Furthermore, NAMP maintenance depends at least in part on humoral signals distinct from FGF-2. In conclusion, our data show a niche/progenitor organization in vitro, in which the BM adherent fraction provides a self-renewing microenvironment for primitive NAMP.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Adult , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Fibroblast Growth Factor 2/genetics , Flow Cytometry , Humans , Middle Aged , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 2/genetics
15.
Biomaterials ; 33(20): 5085-93, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22510434

ABSTRACT

Materials based on synthetic polymers can be extensively tailored in their physical properties but often suffer from limited biological functionality. Here we tested the hypothesis that the biological performance of 3D synthetic polymer-based scaffolds can be enhanced by extracellular matrix (ECM) deposited by cells in vitro and subsequently decellularized. The hypothesis was tested in the context of bone graft substitutes, using polyesterurethane (PEU) foams and mineralized ECM laid by human mesenchymal stromal cells (hMSC). A perfusion-based bioreactor system was critically employed to uniformly seed and culture hMSC in the scaffolds and to efficiently decellularize (94% DNA reduction) the resulting ECM while preserving its main organic and inorganic components. As compared to plain PEU, the decellularized ECM-polymer hybrids supported the osteoblastic differentiation of newly seeded hMSC by up-regulating the mRNA expression of typical osteoblastic genes (6-fold higher bone sialoprotein; 4-fold higher osteocalcin and osteopontin) and increasing calcium deposition (6-fold higher), approaching the performance of ceramic-based materials. After ectopic implantation in nude mice, the decellularized hybrids induced the formation of a mineralized matrix positively immunostained for bone sialoprotein and resembling an immature osteoid tissue. Our findings consolidate the perspective of bioreactor-based production of ECM-decorated polymeric scaffolds as off-the-shelf materials combining tunable physical properties with the physiological presentation of instructive biological signals.


Subject(s)
Biocompatible Materials , Extracellular Matrix , Polymers , Tissue Engineering , Animals , Cell Differentiation , Flow Cytometry , Gene Expression Profiling , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude , Osteoblasts/cytology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Eur Cell Mater ; 21: 445-58; discussion 458, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21604244

ABSTRACT

Three-dimensional (3D) organotypic culture models based on human cells may reduce the use of complex and costly animal models, while gaining clinical relevance. This study aimed at developing a 3D osteoblastic-osteoclastic-endothelial cell co-culture system, as an in vitro model to mimic the process of bone turnover. Osteoprogenitor and endothelial lineage cells were isolated from the stromal vascular fraction (SVF) of human adipose tissue, whereas CD14+ osteoclast progenitors were derived from human peripheral blood. Cells were co-cultured within 3D porous ceramic scaffolds using a perfusion-based bioreactor device, in the presence of typical osteoclastogenic factors. After 3 weeks, the scaffolds contained cells with endothelial (2.0±0.3%), pre/osteoclastic (14.0±1.4%) and mesenchymal/osteoblastic (44.0±8.4%) phenotypes, along with tartrate-resistant acid phosphatase-positive (TRAP+) osteoclastic cells in contact with deposited bone-like matrix. Supernatant analysis demonstrated sustained matrix deposition (by C-terminus procollagen-I propeptides), resorption (by N-terminus collagen-I telopeptides and phosphate levels) and osteoclastic activity (by TRAP-5b) only when SVF and CD14+ cells were co-cultured. Scanning electron microscopy and magnetic resonance imaging confirmed the pattern of matrix deposition and resorption. The effectiveness of Vitamin D in replacing osteoclastogenic factors indicated a functional osteoblast-osteoclast coupling in the system. The formation of human-origin bone-like tissue, blood vessels and osteoclasts upon ectopic implantation validated the functionality of the developed cell types. The 3D co-culture system and the associated non-invasive analytical tools can be used as an advanced model to capture some aspects of the functional coupling of bone-like matrix deposition and resorption and could be exploited toward the engineering of multi-functional bone substitute implants.


Subject(s)
Adult Stem Cells/cytology , Bone and Bones/cytology , Endothelial Cells/metabolism , Acid Phosphatase/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Animals , Antigens, CD/metabolism , Bone Density Conservation Agents/pharmacology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Cell Differentiation , Cell Lineage , Ceramics , Cholecalciferol/pharmacology , Coculture Techniques , Collagen Type I/metabolism , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Humans , Integrin-Binding Sialoprotein/metabolism , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Nude , Monocytes/cytology , Monocytes/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase , Tissue Engineering , Tissue Scaffolds
17.
Proc Natl Acad Sci U S A ; 107(16): 7251-6, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20406908

ABSTRACT

Mesenchymal stem/stromal cells (MSC) are typically used to generate bone tissue by a process resembling intramembranous ossification, i.e., by direct osteoblastic differentiation. However, most bones develop by endochondral ossification, i.e., via remodeling of hypertrophic cartilaginous templates. To date, endochondral bone formation has not been reproduced using human, clinically compliant cell sources. Here, we aimed at engineering tissues from bone marrow-derived, adult human MSC with an intrinsic capacity to undergo endochondral ossification. By analogy to embryonic limb development, we hypothesized that successful execution of the endochondral program depends on the initial formation of hypertrophic cartilaginous templates. Human MSC, subcutaneously implanted into nude mice at various stages of chondrogenic differentiation, formed bone trabeculae only when they had developed in vitro hypertrophic tissue structures. Advanced maturation in vitro resulted in accelerated formation of larger bony tissues. The underlying morphogenetic process was structurally and molecularly similar to the temporal and spatial progression of limb bone development in embryos. In particular, Indian hedgehog signaling was activated at early stages and required for the in vitro formation of hypertrophic cartilage. Subsequent development of a bony collar in vivo was followed by vascularization, osteoclastic resorption of the cartilage template, and appearance of hematopoietic foci. This study reveals the capacity of human MSC to generate bone tissue via an endochondral program and provides a valid model to study mechanisms governing bone development. Most importantly, this process could generate advanced grafts for bone regeneration by invoking a "developmental engineering" paradigm.


Subject(s)
Bone Development , Bone Regeneration , Bone and Bones/pathology , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Transplantation/methods , Chondrocytes/cytology , Hedgehog Proteins/metabolism , Humans , Mice , Regenerative Medicine , Signal Transduction , Tissue Engineering/methods
18.
J Biomed Mater Res A ; 92(4): 1461-7, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19402137

ABSTRACT

In this study, we addressed whether Bone Sialoprotein (BSP) coating of various substrates could enhance the in vitro osteogenic differentiation and in vivo bone formation capacity of human Bone Marrow Stromal Cells (BMSC). Moreover, we tested whether synthetic polymer-based porous scaffolds, despite the absence of a mineral component, could support ectopic bone formation by human BMSC if coated with BSP. Adsorption of recombinant human BSP on tissue culture-treated polystyrene (TCTP), beta-tricalcium phosphate (Osteologic) or synthetic polymer (Polyactive) substrates was dose dependent, but did not consistently accelerate or enhance in vitro BMSC osteogenic differentiation, as assessed by the mRNA expression of osteoblast-related genes. Similarly, BSP coating of porous beta-tricalcium phosphate scaffolds (Skelite) did not improve the efficiency of bone tissue formation following loading with BMSC and ectopic implantation in nude mice. Finally, Polyactive foams seeded with BMSC did not form bone tissue in the same ectopic assay, even if coated with BSP. We conclude that BSP coating of a variety of substrates is not directly associated with an enhancement of osteoprogenitor cell differentiation in vitro or in vivo, and that presentation of BSP on polymeric materials is not sufficient to prime BMSC functional osteoblastic differentiation in vivo.


Subject(s)
Cell Differentiation/drug effects , Ceramics/chemistry , Coated Materials, Biocompatible , Osteogenesis/drug effects , Polymers , Sialoglycoproteins/chemistry , Sialoglycoproteins/pharmacology , Adsorption , Adult , Animals , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Humans , Implants, Experimental , Integrin-Binding Sialoprotein , Materials Testing , Mesenchymal Stem Cell Transplantation , Mice , Mice, Nude , Middle Aged , Osteopontin/genetics , Osteopontin/metabolism , Polymers/chemistry , Polymers/pharmacology , Sialoglycoproteins/genetics , Tissue Scaffolds/chemistry , Tumor Protein, Translationally-Controlled 1
19.
IEEE Trans Neural Netw ; 18(4): 1067-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17668662

ABSTRACT

In this paper, the problem of fault detection in mechanical systems performing linear motion, under the action of friction phenomena is addressed. The friction effects are modeled through the dynamic LuGre model. The proposed architecture is built upon an online neural network (NN) approximator, which requires only system's position and velocity. The friction internal state is not assumed to be available for measurement. The neural fault detection methodology is analyzed with respect to its robustness and sensitivity properties. Rigorous fault detectability conditions and upper bounds for the detection time are also derived. Extensive simulation results showing the effectiveness of the proposed methodology are provided, including a real case study on an industrial actuator.


Subject(s)
Algorithms , Decision Support Techniques , Equipment Failure Analysis/methods , Equipment Failure , Friction , Models, Theoretical , Neural Networks, Computer , Computer Simulation , Mechanics
20.
BMC Bioinformatics ; 8 Suppl 1: S7, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17430574

ABSTRACT

Several systems have been presented in the last years in order to manage the complexity of large microarray experiments. Although good results have been achieved, most systems tend to lack in one or more fields. A Grid based approach may provide a shared, standardized and reliable solution for storage and analysis of biological data, in order to maximize the results of experimental efforts. A Grid framework has been therefore adopted due to the necessity of remotely accessing large amounts of distributed data as well as to scale computational performances for terabyte datasets. Two different biological studies have been planned in order to highlight the benefits that can emerge from our Grid based platform. The described environment relies on storage services and computational services provided by the gLite Grid middleware. The Grid environment is also able to exploit the added value of metadata in order to let users better classify and search experiments. A state-of-art Grid portal has been implemented in order to hide the complexity of framework from end users and to make them able to easily access available services and data. The functional architecture of the portal is described. As a first test of the system performances, a gene expression analysis has been performed on a dataset of Affymetrix GeneChip Rat Expression Array RAE230A, from the ArrayExpress database. The sequence of analysis includes three steps: (i) group opening and image set uploading, (ii) normalization, and (iii) model based gene expression (based on PM/MM difference model). Two different Linux versions (sequential and parallel) of the dChip software have been developed to implement the analysis and have been tested on a cluster. From results, it emerges that the parallelization of the analysis process and the execution of parallel jobs on distributed computational resources actually improve the performances. Moreover, the Grid environment have been tested both against the possibility of uploading and accessing distributed datasets through the Grid middleware and against its ability in managing the execution of jobs on distributed computational resources. Results from the Grid test will be discussed in a further paper.


Subject(s)
Databases, Genetic , Gene Expression Profiling/methods , Information Storage and Retrieval/methods , Models, Biological , Oligonucleotide Array Sequence Analysis/methods , User-Computer Interface , Algorithms , Computer Simulation , Database Management Systems , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...