Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38842036

RESUMO

PURPOSE: Chondrocyte-based cell therapies are effective for the treatment of chondral lesions, but remain poorly indicated for diffuse lesions in the context of early osteoarthritis (OA). The aim of this study was to develop a protocol to obtain chondroprogenitor cells suitable for the treatment of diffuse chondral lesions within early OA. METHODS: Cartilage cells were expanded at low density in human platelet lysate (hPL). A test was performed to exclude senescence. The expression of surface cluster of differentiation 146, cluster of differentiation 166, major histocompatibility complex (MHC)-I and MHC-II and of genes of interest were evaluated, as well as the trophic potential of these cells, by the assessment of lubricin and matrix production. The immunomodulatory potential was assessed through their co-culture with macrophages. RESULTS: Cartilage cells expanded at low density in hPL showed higher proliferation rate than standard-density cells, no replicative senescence, low immunogenicity and expression of lubricin. Moreover, they presented an increased expression of chondrogenic and antihypertrophic markers, as well as a superior matrix deposition if compared to cells cultured at standard density. Cartilage cells induced on macrophages an upregulation of CD206, although a higher increase of CD163 expression was observed in the presence of low-density cells. CONCLUSIONS: These findings lay the grounds to explore the clinical usefulness of low-density cultured cartilage cells to treat diffuse lesions in early OA joints for both autologous and allogenic use. LEVEL OF EVIDENCE: Not applicable.

2.
Front Med (Lausanne) ; 9: 992386, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36314003

RESUMO

The purpose of the present study is to predict by bioinformatics the activity of the extracellular vesicle (EV)-embedded micro RNA (miRNAs) secreted by cartilage cells (CCs), adipose tissue-derived- (ASCs), and bone marrow-derived stem cells (BMSCs) and verify their immunomodulatory potential supporting our bioinformatics findings to optimize the autologous cell-based therapeutic strategies for osteoarthritis (OA) management. Cells were isolated from surgical waste tissues of three patients who underwent total hip replacement, expanded and the EVs were collected. The expression of EV-embedded miRNA was evaluated with the QuantStudio 12 K Flex OpenArray® platform. Mientournet and ingenuity pathway analysis (IPA) were used for validated target prediction analysis and to identify miRNAs involved in OA and inflammation. Cells shared the expression of 325 miRNAs embedded in EVs and differed for the expression of a small number of them. Mienturnet revealed no results for miRNAs selectively expressed by ASCs, whereas miRNA expressed by CCs and BMSCs were putatively involved in the modulation of cell cycle, senescence, apoptosis, Wingless and Int-1 (Wnt), transforming growth factor beta (TGFß), vascular endothelial growth factor (VEGF), Notch, Hippo, tumor necrosis factor alpha (TNFα), interleukin 1 beta (IL-1ß), insulin like growth factor 1 (IGF-1), RUNX family transcription factor 2 (RUNX2), and endochondral ossification pathways. Cartilage homeostasis, macrophages and T cells activity and inflammatory mediators were identified by IPA as targets of the miRNAs found in all the cell populations. Co-culture tests on macrophages and T cells confirmed the immuno-modulatory ability of CCs, ASCs, and BMSCs. The study findings support the rationale behind the use of cell-based therapy for the treatment of OA.

3.
Stem Cell Res Ther ; 13(1): 142, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379348

RESUMO

Mesenchymal stem cell (MSC) culturing for cell therapies needs a step forward to be routinely used in clinical settings. Main concerns regard the use of animal origin reagents, in particular supplementing the culture medium with FBS. Lately, Human Platelet Lysate (HPL) has been proposed as animal-free alternative, described as an excellent supplement for culturing MSCs. The aim of this systematic review was to analyze the current literature on the effect of HPL and FBS on ASCs and BMSCs. The primary outcome was the proliferation rate of cells cultured with FBS and HPL. Differences in terms of doubling time (DT) and population doubling (PD) were evaluated by meta-analysis, subgrouping data according to the cell type. A total of 35 articles were included. BMSCs and ASCs were used in 65.7% (23) and 28.6% (10) studies, respectively. Only two studies included both cell types. Overall, 22 studies were eligible for the meta-analysis. Among them, 9 articles described ASCs and 13 BMSCs. The results showed that BMSCs and ASCs cultured with 10% HPL and 5% HPL have lower DT and higher PD compared to cells cultured with 10% FBS. A possible correlation between the DT decrease and the application of at least 3 freeze/thaw cycles to induce platelet lysis was found. Additionally, HPL increased VEGF secretion and maintained the immuno-modulatory abilities for both cell types. The clarification reported here of the higher efficiency of HPL compared to FBS can help the transition of the scientific community towards clinical-related procedures. 1. The meta-analysis shows that HPL induces a population doubling increase and a doubling time decrease of both ASCs and BMSCs compared to FBS. 2. When at least 3 freeze/thaw cycles are applied to induce platelet lysis, the doubling time of HPL-cultured cells is lower than FBS-cultured cells (Created with BioRender.com).


Assuntos
Plaquetas , Técnicas de Cultura de Células , Células-Tronco Mesenquimais , Soroalbumina Bovina , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Meios de Cultura/metabolismo , Meios de Cultura/farmacologia , Humanos , Células-Tronco Mesenquimais/citologia
4.
Front Bioeng Biotechnol ; 8: 563203, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195126

RESUMO

Osteoarthritis frequently requires arthroplasty. Cementless implants are widely used in clinics to replace damaged cartilage or missing bone tissue. In cementless arthroplasty, the risk of aseptic loosening strictly depends on implant stability and bone-implant interface, which are fundamental to guarantee the long-term success of the implant. Ameliorating the features of prosthetic materials, including their porosity and/or geometry, and identifying osteoconductive and/or osteoinductive coatings of implant surfaces are the main strategies to enhance the bone-implant contact surface area. Herein, the development of a novel composite consisting in the association of macro-porous trabecular titanium with silk fibroin (SF) sponges enriched with anionic fibroin-derived polypeptides is described. This composite is applied to improve early bone ingrowth into the implant mesh in a sheep model of bone defects. The composite enables to nucleate carbonated hydroxyapatite and accelerates the osteoblastic differentiation of resident cells, inducing an outward bone growth, a feature that can be particularly relevant when applying these implants in the case of poor osseointegration. Moreover, the osteoconductive properties of peptide-enriched SF sponges support an inward bone deposition from the native bone towards the implants. This technology can be exploited to improve the biological functionality of various prosthetic materials in terms of early bone fixation and prevention of aseptic loosening in prosthetic surgery.

5.
Nanomedicine (Lond) ; 15(23): 2271-2285, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32914689

RESUMO

Aim: We investigated the use of cellulose nanocrystals (CNCs) as drug nanocarriers combining an anti-osteoporotic agent, alendronate (ALN), and an anti-cancer drug, doxorubicin (DOX). Materials & methods: CNC physicochemical characterization, in vivo imaging coupled with histology and in vitro uptake and toxicity assays were carried out. Results:In vivo CNC-ALN did not modify bone tropism and lung penetration, whereas its liver and kidney accumulation was slightly higher compared with CNCs alone. In vitro studies showed that CNC-ALN did not impair ALN's effect on osteoclasts, whereas CNC-DOX confirmed the therapeutic potential against bone metastatic cancer cells. Conclusions: This study provides robust proof of the potential of CNCs as easy, flexible and specific carriers to deliver compounds to the bone.


Assuntos
Nanopartículas , Preparações Farmacêuticas , Celulose , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos
6.
Artigo em Inglês | MEDLINE | ID: mdl-32984267

RESUMO

Extravasation is a multi-step process implicated in many physiological and pathological events. This process is essential to get leukocytes to the site of injury or infection but is also one of the main steps in the metastatic cascade in which cancer cells leave the primary tumor and migrate to target sites through the vascular route. In this perspective, extravasation is a double-edged sword. This systematic review analyzes microfluidic 3D models that have been designed to investigate the extravasation of cancer and immune cells. The purpose of this systematic review is to provide an exhaustive summary of the advanced microfluidic 3D models that have been designed to study the extravasation of cancer and immune cells, offering a perspective on the current state-of-the-art. To this end, we set the literature search cross-examining PUBMED and EMBASE databases up to January 2020 and further included non-indexed references reported in relevant reviews. The inclusion criteria were defined in agreement between all the investigators, aimed at identifying studies which investigate the extravasation process of cancer cells and/or leukocytes in microfluidic platforms. Twenty seven studies among 174 examined each step of the extravasation process exploiting 3D microfluidic devices and hence were included in our review. The analysis of the results obtained with the use of microfluidic models allowed highlighting shared features and differences in the extravasation of immune and cancer cells, in view of the setup of a common framework, that could be beneficial for the development of therapeutic approaches fostering or hindering the extravasation process.

7.
Cells ; 9(5)2020 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397409

RESUMO

Extracellular vesicles (EVs) showed therapeutic properties in several applications, many in regenerative medicine. A clear example is in the treatment of osteoarthritis (OA), where adipose-derived mesenchymal stem cells (ASCs)-EVs were able to promote regeneration and reduce inflammation in both synovia and cartilage. A still obscure issue is the effective ability of EVs to be internalized by target cells, rather than simply bound to the extracellular matrix (ECM) or plasma membrane, since the current detection or imaging technologies cannot fully decipher it due to technical limitations. In the present study, human articular chondrocytes (ACHs) and fibroblast-like synoviocytes (FLSs) isolated from the same OA patients were cocultured in 2D as well as in 3D conditions with fluorescently labeled ASC-EVs, and analyzed by flow cytometry or confocal microscopy, respectively. In contrast with conventional 2D, in 3D cultures, confocal microscopy allowed a clear detection of the tridimensional morphology of the cells and thus an accurate discrimination of EV interaction with the external and/or internal cell environment. In both 2D and 3D conditions, FLSs were more efficient in interacting with ASC-EVs and 3D imaging demonstrated a faster uptake process. The removal of the hyaluronic acid component from the ECM of both cell types reduced their interaction with ASC-EVs only in the 2D system, showing that 2D and 3D conditions can yield different outcomes when investigating events where ECM plays a key role. These results indicate that studying EVs binding and uptake both in 2D and 3D guarantees a more precise and complementary characterization of the molecular mechanisms involved in the process. The implementation of this strategy can become a valuable tool not only for basic research, but also for release assays and potency prediction for clinical EV batches.


Assuntos
Microambiente Celular , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/citologia , Cartilagem Articular/citologia , Comunicação Celular , Células Cultivadas , Condrócitos/citologia , Endocitose , Feminino , Fibroblastos/citologia , Humanos , Ácido Hialurônico/isolamento & purificação , Dispositivos Lab-On-A-Chip , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Fenótipo , Sinoviócitos/citologia
8.
Curr Opin Biotechnol ; 63: 79-88, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31927146

RESUMO

Multifactorial diseases affecting musculoskeletal tissues are characterized by the interactions between multiple tissues, such as muscle and nerves in neuromuscular diseases, or multiple cellular components in a tissue, as in the case of bone tumors, interacting with bone cells. For these diseases also the influence of different biophysical and biochemical stimuli, such as mechanical overload and inflammatory molecules in osteoarthritis, play a key role. To investigate these complex phenomena, organ-on-a-chip systems have been developed, taking into account specific disease characteristics such as being directly derived from patients, the presence of specifically mutated cells, or a combination of relevant biophysical and/or biochemical stimuli. Depending on the envisaged application, different issues remain to be addressed. In particular, improving automation and output sensors are key for drug screening applications, while refining model microarchitecture to enhance physiological fidelity is needed for more basic science studies.


Assuntos
Dispositivos Lab-On-A-Chip , Doenças Musculoesqueléticas , Avaliação Pré-Clínica de Medicamentos , Humanos , Modelos Biológicos
9.
Stem Cells Int ; 2019: 3715964, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31949437

RESUMO

Nonunions represent one of the major indications for clinical settings with stem cell-based therapies. The objective of this research was to systematically assess the current evidence for the efficacy of bone marrow-derived cell-based approaches associated or not with bone scaffolds for the treatment of nonunions. We searched MEDLINE (PubMed) and CENTRAL up to July 2019 for clinical studies focused on the use of cell-based therapies and bone marrow derivatives to treat bone nonunions. Three investigators independently extracted the data and appraised the risk of bias. We analysed 27 studies including a total number of 347 participants exposed to four interventions: bone marrow concentrate (BMAC), BMAC combined with scaffold (BMAC/Scaffold), bone marrow-derived mesenchymal stromal cells (BMSCs), and BMSC combined with scaffold (BMSC/Scaffold). Two controlled studies showed a positive trend in bone healing in favour of BMAC/Scaffold or BMSC/Scaffold treatment against bone autograft, although the difference was not statistically significant (RR 0.11, 95% CI -0.05; 0.28). Among single cohort studies, the highest mean pooled proportion of healing rate was reported for BMAC (77%; 95% CI 63%-89%; 107 cases, n = 8) and BMAC/Scaffold treatments with (71%; 95% CI 50%-89%; 117 cases, n = 8) at 6 months of follow-up. At 12 months of follow-up, an increasing proportion of bone healing was observed in all the treatment groups, ranging from 81% to 100%. These results indicate that BMAC or BMAC/Scaffold might be considered as the primary choice to treat nonunions with a successful healing rate at a midterm follow-up. Moreover, this meta-analysis highlighted that the presence of a scaffold positively influences the healing rate at a long-term follow-up. More case-control studies are still needed to support the clinical improvement of cell-based therapies against autografts, up to now considered as the gold standard for the treatment of nonunions.

10.
Knee Surg Sports Traumatol Arthrosc ; 27(6): 2003-2020, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30159741

RESUMO

PURPOSE: Osteoarthritis (OA) represents a relevant social and economic burden worldwide. "Mesenchymal stem cells" or, as recently proposed, "medicinal signaling cells" (MSCs) have been recently introduced as injective treatments for OA with the aim of restoring joint homeostasis. The aim of this review is to provide the reader with the tools necessary to interpret the currently available clinical data, focusing on the MSC mechanisms of action which might help to clarify what we should expect from this treatment. METHODS: Clinical studies reporting MSC injections for the treatment of knee OA, either freshly isolated or culture-expanded cells, have been included and commented in relation to the supposed therapeutic effect that MSCs might exert giving their supposed mode of actions. RESULTS: The majority of the studies reports significant improvements in terms of pain and knee function compared to baseline values, up to 24 months of follow-up. Although these data support the expected therapeutic effect of this therapy giving the features of these cells, only 14% of the studies present a control group and more than one-third of them report the results on less than ten patients. CONCLUSIONS: Despite the constant presence of positive and satisfactory results in the studies analyzed, the complexity of MSC metabolism and related therapeutic effects as well as the weakness of most of the studies do not allow withdrawing definitive conclusions about the superiority of one tissue source over another, as well as about the best cell dose and the long-term durability of the effects of these procedures. Given the high potential value of these therapies in the treatment of OA, further studies accurately designed, carefully defining the type of patients to be included and pursuing minimal standard requirements in terms of follow-up, number of patients, and types of measurements should be conducted to finally assess the efficacy of MSC-based injective treatments.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteoartrite do Joelho/terapia , Humanos , Injeções Intra-Articulares , Articulação do Joelho , Osteoartrite do Joelho/fisiopatologia
11.
Differentiation ; 95: 44-53, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28319735

RESUMO

Tendon injuries are severe burdens in clinics. The poor tendon healing is related to an ineffective response of resident cells and inadequate vascularization. Thanks to the high proliferation and multi-lineage differentiation capability, bone marrow-derived mesenchymal stem cells (BMSCs) are a promising cell source to support the tendon repair. To date, the association of various growth factors to induce the in vitro tenogenic differentiation of multipotent progenitor cells is poorly investigated. This study aimed to investigate the tenogenic differentiation of rabbit BMSCs by testing the combination of bone morphogenetic proteins (BMP-12 and 14) with transforming growth factor beta (TGF-ß) and vascular endothelial growth factor (VEGF) both in 2D and 3D cultures within fibrin-based constructs. After 7 and 14 days, the tenogenic differentiation was assessed by analyzing cell metabolism and collagen content, the gene expression of tenogenic markers and the histological cell distribution and collagen deposition within 3D constructs. Our results demonstrated that the association of BMP-14 with TGF-ß3 and VEGF enhanced the BMSC tenogenic differentiation both in 2D and 3D cultures. This study supports the use of fibrin as hydrogel-based matrix to generate spheroids loaded with tenogenic differentiated BMSCs that could be used to treat tendon lesions in the future.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Tendões/citologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/farmacologia , Células Cultivadas , Fibrina/farmacologia , Linfotoxina-alfa/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Coelhos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/farmacologia
12.
Nanomedicine (Lond) ; 11(9): 1073-91, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27078586

RESUMO

AIM: We aimed to establish a 3D vascularized in vitro bone remodeling model. MATERIALS & METHODS: Human umbilical endothelial cells (HUVECs), bone marrow mesenchymal stem cells (BMSCs), and osteoblast (OBs) and osteoclast (OCs) precursors were embedded in collagen/fibrin hydrogels enriched with calcium phosphate nanoparticles (CaPn). We assessed vasculogenesis in HUVEC-BMSC coculture, osteogenesis with OBs, osteoclastogenesis with OCs, and, ultimately, cell interplay in tetraculture. RESULTS: HUVECs developed a robust microvascular network and BMSCs differentiated into mural cells. Noteworthy, OB and OC differentiation was increased by their reciprocal coculture and by CaPn, and even more by the combination of the tetraculture and CaPn. CONCLUSION: We successfully developed a vascularized 3D bone remodeling model, whereby cells interacted and exerted their specific function.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanopartículas/administração & dosagem , Osteogênese/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Fosfatos de Cálcio/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Hidrogéis/administração & dosagem , Hidrogéis/química , Nanopartículas/química , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos
13.
Cell Reprogram ; 17(4): 235-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168053

RESUMO

Bone defects are severe burdens in clinics, and thus cell therapy offers an alternative strategy exploiting the features of bone marrow stromal cells (BMSCs). Sheep are a suitable orthopedic preclinical model for similarities with humans. This study compares the influence of two phosphate sources combined with bone morphogenetic protein-2 (BMP-2) on the osteogenic potential of human and ovine BMSCs. ß-Glycerophosphate (ß-GlyP) and monosodium phosphate (NaH2PO4) were used as organic and inorganic phosphate sources. Osteogenic differentiation of the BMSCs was assessed by calcified matrix, alkaline phosphatase (ALP) activity, and gene expression analysis. A higher calcified matrix deposition was detected in BMSCs cultured with NaH2PO4. Although no significant differences were detected among media for human BMSCs, ß-GlyP with or without BMP-2 determined a positive trend in ALP levels compared to NaH2PO4. In contrast, NaH2PO4 had a positive effect on ALP levels in ovine BMSCs. ß-GlyP better supported the expression of COL1A1 in human BMSCs, whereas all media enhanced RUNX2 and SPARC expression. Ovine BMSCs responded poorly to any media for RUNX2, COL1A1, and SPARC expression. NaH2PO4 improved calcified matrix deposition without upregulating the transcriptional expression of osteogenic markers. A further optimization of differentiation protocols needs to be performed to translate the procedures from preclinical to clinical models.


Assuntos
Glicerofosfatos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fosfatos/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Calcificação Fisiológica , Células Cultivadas , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Feminino , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Osteonectina/genética , Carneiro Doméstico
14.
J Biomed Mater Res B Appl Biomater ; 103(2): 448-56, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24910213

RESUMO

Bone-implant integration represents a major requirement to grant implant stability and reduce the risk of implant loosening. This study investigates the effect of progenitor cells and strontium-enriched hydrogel on the osseointegration of titanium implants. To mimic implant-bone interaction, an ectopic model was developed grafting Trabecular Titanium(™) (TT) implants into decellularized bone seeded with human bone marrow mesenchymal stem cells (hBMSCs). TT was loaded or not with strontium-enriched amidated carboxymethylcellulose (CMCA) hydrogel and/or hBMSCs. Constructs were implanted subcutaneously in athymic mice and osteodeposition was investigated with microcomputed tomography (micro-CT), scanning electron microscopy (SEM), and pull-out test at 4, 8, and 12 weeks. Fluorescence imaging was performed at 8 and 12 weeks, histology at 4 and 8 weeks. Micro-CT demonstrated the homogeneity of the engineered bone in all groups, supporting the reproducibility of the ectopic model. Fluorescence imaging, histology, SEM and pull-out mechanical testing showed superior tissue ingrowth in TT implants loaded with both strontium-enriched CMCA and hBMSCs. In our model, the synergic action of the bioactive hydrogel and hBMSCs increased both the bone deposition and TT integration. Thus, we suggest that using orthopedic prosthetic implant preloaded with strontium-enriched CMCA and seeded with BMSCs could represent a valid single-step surgical strategy to improve implant osseointegration.


Assuntos
Xenoenxertos , Hidrogéis , Implantes Experimentais , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Estrôncio , Titânio , Animais , Células Imobilizadas/metabolismo , Células Imobilizadas/transplante , Feminino , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Masculino , Camundongos , Pessoa de Meia-Idade , Osteogênese , Porosidade , Estrôncio/química , Estrôncio/farmacologia , Titânio/química , Titânio/farmacologia
15.
J Biomed Mater Res A ; 101(12): 3396-403, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23554067

RESUMO

Insufficient implant stability is an important determinant in the failure of cementless prostheses. To improve osseointegration, we aim at generating a bioactive implant combining a macroporous titanium (TT) with a biocompatible hydrogel to encapsulate osteo-inductive factors and osteoprogenitor cells. Amidation and cross-linking degree of an amidated carboxymethylcellulose hydrogel (CMCA) were characterized by FT-IR spectrometry and mechanical testing. Bone marrow mesenchymal stem cells (BMSCs) from osteoarthritic patients were cultured on CMCA hydrogels, TT, and TT loaded with CMCA (TT + CMCA) with an optimized concentration of SrCl2 to evaluate cell viability and osteo-differentiation. Amidation and cross-linking degree were homogeneous among independent CMCA batches. SrCl2 at 5 µg/mL significantly improved BMSCs osteo-differentiation increasing calcified matrix (P < 0.01), type I collagen expression (P < 0.05) and alkaline phosphatase activity. TT + CMCA samples better retained cells into the TT mesh, significantly improving cell seeding efficiency with respect to TT (P < 0.05). BMSCs on TT + CMCA underwent a more efficient osteo-differentiation with higher alkaline phosphatase (P < 0.05) and calcium levels compared to cells on TT. Based on these in vitro results, we envision the association of TT with strontium-enriched CMCA and BMSCs as a promising strategy to generate bioactive implants promoting bone neoformation at the implant site.


Assuntos
Materiais Biocompatíveis/farmacologia , Osso e Ossos/efeitos dos fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Próteses e Implantes , Estrôncio/farmacologia , Titânio/farmacologia , Amidas/química , Carboximetilcelulose Sódica/química , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/farmacologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Ortopedia , Osteogênese/efeitos dos fármacos , Porosidade , Reprodutibilidade dos Testes
16.
Stem Cells Dev ; 19(6): 915-25, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19929314

RESUMO

Stem cells hold great promise in tissue engineering for repairing tissues damaged by disease or injury. Mesenchymal stem cells (MSCs) are multipotent cells able to proliferate and differentiate into multiple mesodermal tissues such as bone, cartilage, muscle, tendon, and fat. We have previously reported that the low-affinity nerve growth factor receptor (L-NGFR or CD271) defines a subset of cells with high proliferative, clonogenic, and multipotential differentiation ability in adult bone marrow (BM). It has been recently shown that adipose tissue is an alternative source of adult multipotent stem cells and human adipose-derived stem cells, selected by plastic adherence (PA hASCs), have been extensively characterized for their functional potentials in vitro. In this study, immunoselected L-NGFR(+) and CD34(+) subpopulations have been analyzed and compared with the PA hASCs. Phenotypic profile of freshly purified subpopulations showed an enrichment in the expression of some stem cell markers; indeed, a great percentage of L-NGFR(+) cells co-expressed CD34 and CD117 antigens, whereas the endothelial-committed progenitor markers KDR and P1H12 were mainly expressed on CD34(+) cells. Differently from PA hASCs, the immunoseparated fractions showed high increments in cell proliferation, and the fibroblast colony-forming activity (CFU-F) was maintained throughout the time of culture. Furthermore, the immunoselected populations showed a greater differentiative potential toward adipocytes, osteoblasts, and chondrocyte-like cells, compared to PA hASCs. Our data suggest that both CD34(+) and L-NGFR(+) hASCs can be considered alternative candidates for tissue engineering and regenerative medicine applications.


Assuntos
Tecido Adiposo/citologia , Anticorpos Monoclonais/imunologia , Antígenos CD34/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Receptor de Fator de Crescimento Neural/imunologia , Adipogenia , Adulto , Biomarcadores/metabolismo , Diferenciação Celular , Membrana Celular/metabolismo , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Matriz Extracelular/metabolismo , Feminino , Glicosaminoglicanos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/metabolismo , Frações Subcelulares/metabolismo
17.
Cell Tissue Res ; 338(3): 401-11, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19882172

RESUMO

One of the most important issues in orthopaedic surgery is the loss of bone resulting from trauma, infections, tumours or congenital deficiency. In view of the hypothetical future application of mesenchymal stem cells isolated from human adipose tissue in regenerative medicine, we have analysed and characterized adipose-derived stem cells (ASCs) isolated from adipose tissue of rat, rabbit and pig. We have compared their in vitro osteogenic differentiation abilities for exploitation in the repair of critical osteochondral defects in autologous pre-clinical models. The number of pluripotent cells per millilitre of adipose tissue is variable and the yield of rabbit ASCs is lower than that in rat and pig. However, all ASCs populations show both a stable doubling time during culture and a marked clonogenic ability. After exposure to osteogenic stimuli, ASCs from rat, rabbit and pig exhibit a significant increase in the expression of osteogenic markers such as alkaline phosphatase, extracellular calcium deposition, osteocalcin and osteonectin. However, differences have been observed depending on the animal species and/or differentiation period. Rabbit and porcine ASCs have been differentiated on granules of clinical grade hydroxyapatite (HA) towards osteoblast-like cells. These cells grow and adhere to the scaffold, with no inhibitory effect of HA during osteo-differentiation. Such in vitro studies are necessary in order to select suitable pre-clinical models to validate the use of autologous ASCs, alone or in association with proper biomaterials, for the repair of critical bone defects.


Assuntos
Tecido Adiposo/citologia , Osteogênese , Células-Tronco/citologia , Animais , Materiais Biocompatíveis/química , Proliferação de Células , Separação Celular , Sobrevivência Celular , Células Cultivadas , Durapatita/química , Humanos , Coelhos , Ratos , Suínos , Alicerces Teciduais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA