Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Lancet Neurol ; 21(6): 528-536, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35568047

RESUMO

BACKGROUND: Perinatal arterial ischaemic stroke (PAIS) is an important cause of neurodevelopmental disabilities. In this first-in-human study, we aimed to assess the feasibility and safety of intranasally delivered bone marrow-derived allogeneic mesenchymal stromal cells (MSCs) to treat PAIS in neonates. METHODS: In this open-label intervention study in collaboration with all neonatal intensive care units in the Netherlands, we included neonates born at full term (≥36 weeks of gestation) with MRI-confirmed PAIS in the middle cerebral artery region. All eligible patients were transferred to the neonatal intensive care unit of the Wilhelmina Children's Hospital. Neonates received one dose of 45-50 × 106 bone-marrow derived MSCs intranasally within 7 days of presenting signs of PAIS. The primary endpoints were acute and subacute safety outcomes, including vital signs, blood markers, and the occurrence of toxicity, adverse events, and serious adverse events. The occurrence of unexpected cerebral abnormalities by a repeat MRI at 3 months of age was a secondary endpoint. As part of standard clinical follow-up at Wilhelmina Children's Hospital, we assessed corticospinal tract development on MRI and performed motor assessments at 4 months of age. This study is registered with ClinicalTrials.gov, NCT03356821. FINDINGS: Between Feb 11, 2020, and April 29, 2021, ten neonates were enrolled in the study. Intranasal administration of MSCs was well tolerated in all ten neonates. No serious adverse events were observed. One adverse event was seen: a mild transient fever of 38°C without the need for clinical intervention. Blood inflammation markers (C-reactive protein, procalcitonin, and leukocyte count) were not significantly different pre-administration versus post-administration and, although thrombocyte levels increased (p=0·011), all were within the physiological range. Follow-up MRI scans did not show unexpected structural cerebral abnormalities. All ten patients had initial pre-Wallerian changes in the corticospinal tracts, but only four (40%) patients showed asymmetrical corticospinal tracts at follow-up MRI. Abnormal early motor assessment was found in three (30%) infants. INTERPRETATION: This first-in-human study demonstrates that intranasal bone marrow-derived MSC administration in neonates after PAIS is feasible and no serious adverse events were observed in patients followed up until 3 months of age. Future large-scale placebo-controlled studies are needed to determine the therapeutic effect of intranasal MSCs for PAIS. FUNDING: Netherlands Organization for Health Research and Development (ZonMw).


Assuntos
Isquemia Encefálica , AVC Isquêmico , Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Criança , Estudos de Viabilidade , Humanos , Lactente , Recém-Nascido , Países Baixos , Pesquisa , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/terapia , Resultado do Tratamento
2.
Stem Cell Reports ; 14(2): 300-311, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31956083

RESUMO

RAG2 severe combined immune deficiency (RAG2-SCID) is a lethal disorder caused by the absence of functional T and B cells due to a differentiation block. Here, we generated induced pluripotent stem cells (iPSCs) from a RAG2-SCID patient to study the nature of the T cell developmental blockade. We observed a strongly reduced capacity to differentiate at every investigated stage of T cell development, from early CD7-CD5- to CD4+CD8+. The impaired differentiation was accompanied by an increase in CD7-CD56+CD33+ natural killer (NK) cell-like cells. T cell receptor D rearrangements were completely absent in RAG2SCID cells, whereas the rare T cell receptor B rearrangements were likely the result of illegitimate rearrangements. Repair of RAG2 restored the capacity to induce T cell receptor rearrangements, normalized T cell development, and corrected the NK cell-like phenotype. In conclusion, we succeeded in generating an iPSC-based RAG2-SCID model, which enabled the identification of previously unrecognized disorder-related T cell developmental roadblocks.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Linfócitos T/imunologia , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem da Célula , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Hematopoese , Humanos , Células Matadoras Naturais/imunologia , Camundongos SCID
4.
Stem Cells Transl Med ; 5(10): 1362-1374, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27388241

RESUMO

: In patients undergoing maxillary sinus floor elevation (MSFE) for dental implant placement, bone substitutes are currently evaluated as alternatives for autologous bone. However, bone substitutes have only osteoconductive properties and lack osteoinductive potential. Therefore, this phase I study evaluated the potential additive effect on bone regeneration by the addition of freshly isolated, autologous but heterologous stromal vascular fraction (SVF), which is highly enriched with adipose stromal/stem cells when compared with native adipose tissue. From 10 patients, SVF was procured using automatic processing, seeded on either ß-tricalcium phosphate (n = 5) or biphasic calcium phosphate carriers (n = 5), and used for MSFE in a one-step surgical procedure. Primary objectives were feasibility and safety. The secondary objective was efficacy, evaluated by using biopsies of the augmented area taken 6 months postoperatively, concomitant with dental implant placement. Biopsies were assessed for bone, graft, and osteoid volumes. No adverse effects were reported during the procedure or follow-up (≥3 years). Bone and osteoid percentages were higher in study biopsies (SVF supplemented) than in control biopsies (ceramic only on contralateral side), in particular in ß-tricalcium phosphate-treated patients. Paired analysis on the six bilaterally treated patients revealed markedly higher bone and osteoid volumes using microcomputed tomography or histomorphometric evaluations, demonstrating an additive effect of SVF supplementation, independent of the bone substitute. This study demonstrated for the first time the feasibility, safety, and potential efficacy of SVF seeded on bone substitutes for MSFE, providing the first step toward a novel treatment concept that might offer broad potential for SVF-based regenerative medicine applications. SIGNIFICANCE: This is the first-in-human study using freshly isolated, autologous adipose stem cell preparations (the stromal vascular fraction [SVF] of adipose tissue) applied in a one-step surgical procedure with calcium phosphate ceramics (CaP) to increase maxillary bone height for dental implantations. All 10 patients received CaP plus SVF on one side, whereas bilaterally treated patients (6 of 10) received CaP only on the opposite side. This allowed intrapatient evaluation of the potential added value of SVF supplementation, assessed in biopsies obtained after 6 months. Feasibility, safety, and potential efficacy of SVF for bone regeneration were demonstrated, showing high potential for this novel concept.


Assuntos
Regeneração Óssea , Fosfatos de Cálcio/uso terapêutico , Transplante de Células-Tronco Mesenquimais , Levantamento do Assoalho do Seio Maxilar/métodos , Tecido Adiposo/citologia , Idoso , Implantes Dentários , Feminino , Citometria de Fluxo , Humanos , Masculino , Maxila , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Medicina Regenerativa/métodos , Microtomografia por Raio-X
5.
J Immunother ; 39(2): 90-100, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26849078

RESUMO

Cell-based immunotherapy using donor-derived natural killer (NK) cells after allogeneic hematopoietic stem cell transplantation may be an attractive treatment of residual leukemia. This study aimed to optimize clinical grade production of a cytokine-activated NK-cell product. NK cells were isolated either by double depletion (CD3(-), CD19(-)) or by sequential depletion and enrichment (CD3(-,) CD56(+)) via CliniMACS from leukapheresis material and cultured in vitro with interleukin (IL)-2 or IL-15. Both NK cell isolation procedures yielded comparable recovery of NK cells and levels of T-cell contamination. After culture with cytokines, the CD3(-)CD56(+) procedure resulted in NK cells of higher purity, that is, less T cells and monocytes, higher viability, and a slightly higher yield than the CD3(-)CD19- procedure. CD69, NKp44, and NKG2A expression were higher on CD3(-)CD56(+) products, whereas lysis of Daudi cells was comparable. Five days of culture led to higher expression of CD69, NKp44, and NKp30 and lysis of K562 and Daudi cell lines. Although CD69 expression and lysis of Daudi cells were slightly higher in cultures with IL-2, T-cell contamination was lower with IL-15. Therefore, further experiments were performed with CD3(-)CD56(+) products cultured with IL-15. Cryopreservation of IL-15-activated NK cells resulted in a loss of cytotoxicity (>92%), whereas thawing of isolated, uncultured NK cells followed by culture with IL-15 yielded cells with about 43% of the original lytic activity. Five-day IL-15-activated NK cells lysed tumor target cell lines and primary leukemic blasts, providing the basis for NK cell­based immunotherapeutic strategies in a clinical setting.


Assuntos
Células Matadoras Induzidas por Citocinas/imunologia , Transplante de Células-Tronco Hematopoéticas , Células Matadoras Naturais/imunologia , Leucemia/terapia , Antígenos CD/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Criopreservação , Células Matadoras Induzidas por Citocinas/transplante , Citocinas/metabolismo , Citotoxicidade Imunológica , Humanos , Células Matadoras Naturais/transplante , Leucemia/imunologia
6.
Tissue Eng Part A ; 22(3-4): 336-48, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26732248

RESUMO

Partial replacement of chondrocytes by stem cells has been proposed to improve the performance of autologous chondrocyte implantation (ACI). Our previous studies showed that the increased cartilage production in pellet cocultures of chondrocytes and mesenchymal stem cells (MSCs) is due to a trophic role of the MSCs by stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. The aim of this study is to compare the trophic effects of stromal vascular fraction cells (SVF) and in vitro expanded adipose stem cells (ASC). SVF and culture-expanded ASC (n = 9) were cocultured with primary human chondrocytes in pellets. By glycosaminoglycan (GAG) and DNA assays, we showed that coculture pellets of SVF and chondrocytes have more GAG deposition than that of ASC and chondrocytes. Results of the short tandem repeats analysis indicated that the increase in the chondrocyte proportion in the coculture pellets is more pronounced in the SVF coculture group than in the ASC coculture group. Using flow cytometry and microarray, we demonstrated that SVF and ASC have different characteristics in cell surface markers and gene expression profiles. SVF is more heterogeneous than ASC, whereas ASC is more enriched in cells from the mesenchymal lineage than SVF. By subcutaneous implantation into nude mice, we showed that constructs of SVF and chondrocytes are better in depositing cartilage matrix than the mixture of ASC and chondrocytes. Taken together, SVF is better than ASC in terms of forming cartilage matrix in pellet coculture and in coimplantation models omitting the need for prior cell expansion. Our study suggests that the SVF in combination with primary human chondrocytes may be a good cell combination for one-stage cartilage repair.


Assuntos
Tecido Adiposo/metabolismo , Antígenos de Diferenciação/biossíntese , Cartilagem/metabolismo , Condrócitos/metabolismo , Condrogênese , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/citologia , Animais , Cartilagem/citologia , Condrócitos/citologia , Técnicas de Cocultura , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos
7.
J Tissue Eng Regen Med ; 10(3): 233-44, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255230

RESUMO

The combination of scaffolds and mesenchymal stromal cells (MSCs) is a promising approach in bone tissue engineering (BTE). Knowledge on the survival, outgrowth and bone-forming capacity of MSCs in vivo is limited. Bioluminescence imaging (BLI), histomorphometry and immunohistochemistry were combined to study the fate of gene-marked goat and human MSCs (gMSCs, hMSCs) on scaffolds with different osteoinductive properties. Luciferase-GFP-labelled MSCs were seeded on hydroxyapatite (HA) or ß-tricalcium phosphate (TCP), cultured for 7 days in vitro in osteogenic medium, implanted subcutaneously in immunodeficient mice and monitored with BLI for 6 weeks. The constructs were retrieved and processed for histomorphometry and detection of luciferase-positive cells (LPCs). For gMSCs, BLI revealed doubling of signal after 1 week, declining to 60% of input after 3 weeks and remaining constant until week 6. hMSCs showed a constant decrease of BLI signal to 25% of input, indicating no further expansion. Bone formation of gMSCs was two-fold higher on TCP than HA. hMSCs and gMSCs control samples produced equal amounts of bone on TCP. Upon transduction, there was a four-fold reduction in bone formation compared with untransduced hMSCs, and no bone was formed on HA. LPCs were detected at day 14, but were much less frequent at day 42. Striking differences were observed in spatial distribution. MSCs in TCP were found to be aligned and interconnected on the surface but were scattered in an unstructured fashion in HA. In conclusion, the spatial distribution of MSCs on the scaffold is critical for cell-scaffold-based BTE.


Assuntos
Fosfatos de Cálcio/farmacologia , Durapatita/farmacologia , Células-Tronco Mesenquimais/citologia , Animais , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Cabras , Humanos , Imuno-Histoquímica , Luciferases/metabolismo , Medições Luminescentes , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos BALB C , Osteogênese/efeitos dos fármacos , Alicerces Teciduais/química
8.
J Biomed Mater Res A ; 103(2): 439-50, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24737694

RESUMO

The aim of this study was to compare the effect of implantation site (i.e., subcutaneous, SQ vs. intramuscular, IM) on bone forming capacity of cell-based and growth factor-based scaffolds in athymic nude rats after an implantation period of 8 weeks. Cell-based scaffolds consisted of porous hydroxyapatite/tricalcium phosphate (HA/TCP) scaffolds seeded with either human adipose tissue-derived mesenchymal stem cells (AT-MSCs) only or both AT-MSCs and human umbilical vein endothelial cells (HUVECs), which were precultured in osteogenic medium for 7 days. Growth factor-based scaffolds consisted of porous HA/TCP scaffolds with 20 µg preadsorbed bone morphogenetic protein-2 (BMP-2). Histological and histomorphometrical analysis were used to assess bone formation. A differentiation experiment was performed in parallel to compare the in vitro osteogenic capacity of cell-based scaffolds. The results showed that cell-based scaffolds showed evident osteogenic differentiation in vitro, with only marginal differences between AT-MSCs only and AT-MSCs/HUVECs. In vivo, none of the cell-based scaffolds showed bone formation, irrespective of the site of implantation. In contrast, all growth factor-based scaffolds showed bone formation at both implantation sites without differences in the amount of formed bone. In conclusion, the results of this study demonstrated that the bone forming capacity of HA/TCP scaffolds with pre-adsorbed BMP-2 was equal at different ectopic implantation sites. Further, despite obvious in vitro osteogenic differentiation of AT-MSCs and AT-MSCS/HUVECs on HA/TCP scaffolds, no bone formation of these cell-based scaffolds was observed in vivo. This indicates further investigation on bone formation mechanisms of AT-MSCs is needed before AT-MSCs can be used as a cytotherapeutic treatment in clinics.


Assuntos
Fosfatos de Cálcio/química , Diferenciação Celular , Durapatita/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Implantes Experimentais , Osteogênese , Alicerces Teciduais/química , Animais , Proteína Morfogenética Óssea 2/química , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Masculino , Ratos , Ratos Nus
9.
J Tissue Eng Regen Med ; 9(7): 779-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23364774

RESUMO

Human bone marrow-derived mesenchymal stem cells (BM-MSCs) and human adipose tissue-derived mesenchymal stem cells (AT-MSCs) are the most frequently used stem cells in tissue engineering. Due to major clinical demands, it is necessary to find an optimally safe and efficient way for large-scale expansion of these cells. Considering the nutritional source in the culture medium and method, this study aimed to analyze the effects of FBS- and PL-supplemented media on osteogenesis in stem cell mono- and co-cultures with human umbilical vein endothelial cells (HUVECs). Results showed that cell metabolic activity and proliferation increased in PL- compared to FBS-supplemented media in mono- and co-cultures for both BM-MSCs and AT-MSCs. In addition, calcium deposition was cell type dependent and decreased for BM-MSCs but increased for AT-MSCs in PL-supplemented medium in both mono- and co-cultures. Based on the effects of co-cultures, BM-MSCs/HUVECs enhanced osteogenesis compared to BM-MSCs monocultures in both FBS- and PL-supplemented media whereas AT-MSCs/HUVECs showed similar results compared to AT-MSCs monocultures.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Técnicas de Cocultura/métodos , Meios de Cultura , Células Endoteliais da Veia Umbilical Humana/citologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/metabolismo , Células da Medula Óssea/metabolismo , Meios de Cultura/química , Meios de Cultura/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo
10.
Biofabrication ; 6(1): 015005, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24429700

RESUMO

The aim of this study was to comparatively evaluate the angiogenic capacity of cocultures using either human bone marrow- or human adipose tissue-derived mesenchymal stem cells (MSCs) (BM- or AT-MSCs) with human umbilical vein endothelial cells (HUVECs) both in vitro and in vivo at early time points (i.e. days 3 and 7). In vitro, cells were either monocultured (i.e. BM-MSCs, AT-MSCs or HUVECs) or cocultured (i.e. BM-MSCs/HUVECs and AT-MSCs/HUVECs) on Thermanox® (2-dimensional, 2D) or in collagen gels (3-dimensional, 3D). For the in vivo experiment, cells (cocultures) were embedded in collagen gels and implanted subcutaneously in nude mice. For both in vitro and in vivo experiments, samples were collected on days 3 and 7 and histologically processed for hematoxylin-eosin and platelet endothelial cell adhesion molecule (PECAM-1; CD31) staining. For in vivo samples, quantitative parameters for evaluating angiogenesis included CD31-positive staining percentage, total vessel-like structure (VLS) area percentage, VLS density, and average VLS area (i.e. the size of per VLS). In vitro results showed the formation of VLS in both cocultures, while none of the monocultures showed VLS formation, irrespective of 2D or 3D culture condition. Although VLS formation occurred after in vivo implantation, no significant difference in angiogenic capacity was observed between the two cocultures, either on day 3 or on day 7. Further, VLS density decreased and anastomosis of the new human vessels with the murine host vasculature occurred over time. In conclusion, this study demonstrated that AT-MSCs/HUVECs and BM-MSCs/HUVECs have equal angiogenic capacity both in vitro and in vivo, and that vessels from donor origin can anastomose with the host vasculature within seven days of implantation.


Assuntos
Tecido Adiposo/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Células da Medula Óssea/fisiologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Engenharia Tecidual , Tecido Adiposo/metabolismo , Adulto , Animais , Vasos Sanguíneos/metabolismo , Células da Medula Óssea/citologia , Células Cultivadas , Técnicas de Cocultura , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Engenharia Tecidual/métodos
11.
Stem Cell Res ; 12(2): 428-40, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24384458

RESUMO

One of the applications of bone marrow stromal cells (BMSCs) that are produced by ex vivo expansion is for use in in vivo bone tissue engineering. Cultured stromal cells are a mixture of cells at different stages of commitment and expansion capability, leading to a heterogeneous cell population that each time can differ in the potential to form in vivo bone. A parameter that predicts for in vivo bone forming capacity is thus far lacking. We employed single colony-derived BMSC cultures to identify such predictive parameters. Using limiting dilution, we have produced sixteen single CFU-F derived BMSC cultures from human bone marrow and found that only five of these formed bone in vivo. The single colony-derived BMSC strains were tested for proliferation, osteogenic-, adipogenic- and chondrogenic differentiation capacity and the expression of a variety of associated markers. The only robust predictors of in vivo bone forming capacity were the induction of alkaline phosphatase, (ALP) mRNA levels and ALP activity during in vitro osteogenic differentiation. The predictive value of in vitro ALP induction was confirmed by analyzing "bulk-cultured" BMSCs from various bone marrow biopsies. Our findings show that in BMSCs, the additional increase in ALP levels over basal levels during in vitro osteogenic differentiation is predictive of in vivo performance.


Assuntos
Fosfatase Alcalina/metabolismo , Células da Medula Óssea/enzimologia , Células-Tronco Mesenquimais/enzimologia , Osteogênese/fisiologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Células Cultivadas , Criança , Indução Enzimática , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Engenharia Tecidual
12.
J Biomed Mater Res A ; 102(4): 1026-36, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23640784

RESUMO

The aim of this study was to compare the osteogenic capacity between human adipose tissue-derived mesenchymal stem cells (AT-MSCs) and their cocultures with human umbilical vein endothelial cells (HUVECs) in vitro and their biological performance in vivo. First, the optimal cell ratio in cocultures for osteogenic differentiation was determined by seeding AT-MSCs and HUVECs in ratios varying from 100:0 to 0:100 on tissue culture plates. Afterward, AT-MSCs and AT-MSCs/HUVECs (50:50) were seeded on porous titanium fiber mesh scaffolds (Ti) for both in vitro and in vivo osteogenic evaluation. For in vitro evaluation, cell osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity and calcium assay. For in vivo evaluation, the scaffolds were implanted bilaterally into rat cranial defects (5 mm diameter) and bone formation was assessed histologically and histomorphometrically after 8 weeks. The ratio of 50:50 was chosen in the cocultures because this coculture condition retained similar amount of calcium deposition while using the least amount of AT-MSCs. Moreover, AT-MSCs showed higher osteogenic differentiation in comparison to AT-MSCs/HUVECs on Ti in vitro. Furthermore, superior bone formation was observed in AT-MSCs compared to AT-MSCs/HUVECs in rat cranial defects. In conclusion, AT-MSCs showed significantly higher osteogenic potential compared to AT-MSCs/HUVECs both in vitro and in vivo.


Assuntos
Tecido Adiposo/citologia , Células Endoteliais da Veia Umbilical Humana/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Crânio/patologia , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Implantação de Prótese , Ratos , Ratos Nus , Crânio/diagnóstico por imagem , Crânio/efeitos dos fármacos , Alicerces Teciduais/química , Titânio/farmacologia , Microtomografia por Raio-X
13.
Eur Cell Mater ; 25: 215-28, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23529785

RESUMO

Calcium phosphates are used in maxillary sinus floor elevation (MSFE) procedures to increase bone height prior to dental implant placement. Whether a collagenous barrier membrane coverage of the lateral window affects bone formation within a bone substitute augmentation is currently an important matter of debate, since its benefit has not been irrefutably proven. Therefore, in this clinical study twelve patients underwent an MSFE procedure with ß-tricalcium phosphate (ß-TCP). The lateral window was either left uncovered, or covered with a resorbable collagenous barrier membrane. After a 6-months healing period, bone biopsies were retrieved during implant placement. Consecutive 1 mm regions of interest of these biopsies were assessed for bone formation, resorption parameters, as well as bone architecture using histology, histomorphometry and micro-computed tomography. Comparable outcomes between the groups with and without membrane were observed regarding osteoconduction rate, bone and graft volume, osteoclast number and structural parameters of newly formed bone per region of interest. However, osteoid volume in grafted maxillary sinus floors without membrane was significantly higher than with membrane. In conclusion, our results - obtained with a novel method employed using 1 mm regions of interest - demonstrate that the clinical application of a bioresorbable collagenous barrier membrane covering the lateral window, after an MSFE procedure with ß-TCP, was not beneficial for bone regeneration and even decreased osteoid production which might lead to diminished bone formation in the long run.


Assuntos
Substitutos Ósseos/uso terapêutico , Fosfatos de Cálcio/uso terapêutico , Colágeno/uso terapêutico , Levantamento do Assoalho do Seio Maxilar , Perda de Dente/cirurgia , Implantes Absorvíveis , Adulto , Idoso , Regeneração Óssea , Feminino , Humanos , Masculino , Maxila/diagnóstico por imagem , Maxila/patologia , Maxila/cirurgia , Membranas Artificiais , Pessoa de Meia-Idade , Osteoclastos/efeitos dos fármacos , Perda de Dente/diagnóstico por imagem , Perda de Dente/patologia , Resultado do Tratamento , Microtomografia por Raio-X
14.
Tissue Eng Part B Rev ; 19(1): 69-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22909124

RESUMO

Bone loss in the oral and maxillofacial region caused by trauma, tumors, congenital disorders, or degenerative diseases is a health care problem worldwide. To restore (reconstruct) these bone defects, human or animal bone grafts or alloplastic (synthetic) materials have been used. However, several disadvantages are associated with bone graft transplantation, such as limited bone volume, donor-site morbidity, surgical and immune rejection risks, and lack of osseo-integration. Bone tissue engineering is emerging as a valid alternative to treat bone defects allowing the regeneration of lost bony tissue, thereby recovering its functionality. During the last decades, the increasing aged population worldwide has also raised the prevalence of maxillary atrophy. Maxillary sinus floor elevation (MSFE) has become a standard surgical procedure to overcome the reduced amount of bone, thus enabling the placement of dental implants. MSFE aims to increase the bone height in the posterior maxilla, by elevating the Schneiderian membrane and placing the graft material into the surgically created space in the maxillary sinus floor. Importantly, oral bone regeneration during MSFE offers a unique human clinical model in which new cell-based bone tissue engineering applications might be investigated, since biopsies can be taken after MSFE before a dental implant placement and analyzed at the cellular level. New approaches in oral bone regeneration are focusing on cells, growth factors, and biomaterials. Recently, adipose tissue has become interesting as an abundant source of mesenchymal stem cells, which might be applied immediately after isolation to the patient allowing a one-step surgical procedure, thereby avoiding expensive cell culture procedures and another surgical operation. In this new cell-based tissue engineering approach, stem cells are combined with an osteoconductive scaffold and growth factors, and applied immediately to the patient. In this review, MSFE is discussed as a valid model to test bone tissue engineering approaches, such as the one-step surgical procedure. This procedure might be applied in other regenerative medicine applications as well.


Assuntos
Regeneração Óssea , Substitutos Ósseos/uso terapêutico , Regeneração Tecidual Guiada/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Levantamento do Assoalho do Seio Maxilar/instrumentação , Levantamento do Assoalho do Seio Maxilar/métodos , Humanos
15.
Stem Cells Dev ; 22(7): 1086-96, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23145933

RESUMO

Bone marrow (BM) stromal cells (MSCs), also known as mesenchymal stem cells, display a high degree of heterogeneity. To shed light on the causes of this heterogeneity, MSCs were collected from either human BM (n=5) or adipose tissue (AT) (n=5), and expanded using 2 different culture methods: one based on fetal calf serum, and one based on human platelet lysate. After initial expansion, MSCs were frozen, and the vials were transported to 3 different laboratories and grown for 1 passage using the same brand of culture plastic, medium, and supplements. Subsequently, the cells were harvested and assayed for their gene expression profile using the Affymetrix exon microarray platform. Based on gene expression profiles, the most discriminative feature was the anatomical harvesting site, followed by culture methodology. Remarkably, genes in the WNT pathway were expressed at higher levels in BM-derived MSCs than in AT-derived MSCs. Although differences were found between laboratories, cell culture location only slightly affects heterogeneity. Furthermore, individual donors contributed marginally to the observed differences in transcriptomes. Finally, BM-derived MSCs displayed the highest level of similarity, irrespective their culture conditions, when compared to AT-derived cells.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Tecido Adiposo/metabolismo , Células da Medula Óssea/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt/genética
16.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-22653974

RESUMO

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Nicho de Células-Tronco/imunologia , Quimeras de Transplante/imunologia , Microambiente Tumoral/imunologia , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Ossículos da Orelha/citologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Camundongos , Camundongos Mutantes , Transplante de Neoplasias , Osteólise/imunologia , Alicerces Teciduais , Transplante Heterólogo
17.
Tissue Eng Part A ; 18(15-16): 1542-51, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22429306

RESUMO

Earlier, we have shown that the increased cartilage production in pellet co-cultures of chondrocytes and bone marrow-derived mesenchymal stem cells (BM-MSCs) is due to a trophic role of the MSC in stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. These studies were performed in a culture medium that was not compatible with the chondrogenic differentiation of MSCs. In this study, we tested whether the trophic role of the MSCs is dependent on culturing co-culture pellets in a medium that is compatible with the chondrogenic differentiation of MSCs. In addition, we investigated whether the trophic role of the MSCs is dependent on their origins or is a more general characteristic of MSCs. Human BM-MSCs and bovine primary chondrocytes were co-cultured in a medium that was compatible with the chondrogenic differentiation of MSCs. Enhanced matrix production was confirmed by glycosaminoglycans (GAG) quantification. A species-specific quantitative polymerase chain reaction demonstrated that the cartilage matrix was mainly of bovine origin, indicative of a lack of the chondrogenic differentiation of MSCs. In addition, pellet co-cultures were overgrown by bovine cells over time. To test the influence of origin on MSCs' trophic effects, the MSCs isolated from adipose tissue and the synovial membrane were co-cultured with human primary chondrocytes, and their activity was compared with BM-MSCs, which served as control. GAG quantification again confirmed increased cartilage matrix production, irrespective of the source of the MSCs. EdU staining combined with cell tracking revealed an increased proliferation of chondrocytes in each condition. Irrespective of the MSC source, a short tandem repeat analysis of genomic DNA showed a decrease in MSCs in the co-culture over time. Our results clearly demonstrate that in co-culture pellets, the MSCs stimulate cartilage formation due to a trophic effect on the chondrocytes rather than differentiating into chondrocytes, irrespective of culture condition or origin. This implies that the trophic effect of MSCs in co-cultures is a general phenomenon with potential implications for use in cartilage repair strategies.


Assuntos
Técnicas de Cultura de Células/métodos , Condrócitos/citologia , Células-Tronco Mesenquimais/citologia , Animais , Bovinos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultura/farmacologia , Matriz Extracelular/metabolismo , Glicosaminoglicanos/biossíntese , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo
18.
Tissue Eng Part C Methods ; 18(1): 33-44, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21854293

RESUMO

Osteochondral defects are prone to induce osteoarthritic degenerative changes. Many tissue-engineering approaches that aim to generate osteochondral implants suffer from poor tissue formation and compromised integration. This illustrates the need for further improvement of heterogeneous tissue constructs. Engineering of these structures is expected to profit from strategies addressing the complexity of tissue organization and the simultaneous use of multiple cell types. Moreover, this enables the investigation of the effects of three-dimensional (3D) organization and architecture on tissue function. In the present study, we characterize the use of a 3D fiber deposition (3DF) technique for the fabrication of cell-laden, heterogeneous hydrogel constructs for potential use as osteochondral grafts. Changing fiber spacing or angle of fiber deposition yielded scaffolds of varying porosity and elastic modulus. We encapsulated and printed fluorescently labeled human chondrocytes and osteogenic progenitors in alginate hydrogel yielding scaffolds of 1×2 cm with different parts for both cell types. Cell viability remained high throughout the printing process, and cells remained in their compartment of the printed scaffold for the whole culture period. Moreover, distinctive tissue formation was observed, both in vitro after 3 weeks and in vivo (6 weeks subcutaneously in immunodeficient mice), at different locations within one construct. These results demonstrate the possibility of manufacturing viable centimeter-scaled structured tissues by the 3DF technique, which could potentially be used for the repair of osteochondral defects.


Assuntos
Condrócitos/citologia , Hidrogéis/química , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Sobrevivência Celular , Feminino , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Microscopia de Fluorescência/métodos , Modelos Biológicos , Porosidade , Estresse Mecânico , Viscosidade
19.
Cancer Cell ; 20(3): 370-83, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21907927

RESUMO

The development of resistance to chemotherapy is a major obstacle for lasting effective treatment of cancer. Here, we demonstrate that endogenous mesenchymal stem cells (MSCs) become activated during treatment with platinum analogs and secrete factors that protect tumor cells against a range of chemotherapeutics. Through a metabolomics approach, we identified two distinct platinum-induced polyunsaturated fatty acids (PIFAs), 12-oxo-5,8,10-heptadecatrienoic acid (KHT) and hexadeca-4,7,10,13-tetraenoic acid (16:4(n-3)), that in minute quantities induce resistance to a broad spectrum of chemotherapeutic agents. Interestingly, blocking central enzymes involved in the production of these PIFAs (cyclooxygenase-1 and thromboxane synthase) prevents MSC-induced resistance. Our findings show that MSCs are potent mediators of resistance to chemotherapy and reveal targets to enhance chemotherapy efficacy in patients.


Assuntos
Antineoplásicos/farmacologia , Ciclo-Oxigenase 1/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ácidos Graxos Insaturados/metabolismo , Ácidos Graxos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Compostos de Platina/farmacologia , Tromboxano-A Sintase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carboplatina/administração & dosagem , Carboplatina/farmacologia , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Inibidores de Ciclo-Oxigenase , Humanos , Espectrometria de Massas , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Tromboxano-A Sintase/antagonistas & inibidores , Células Tumorais Cultivadas
20.
Tissue Eng Part A ; 16(11): 3343-51, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20575656

RESUMO

Tissue engineering of bone, by combining multipotent stromal cells (MSCs) with osteoconductive scaffolds, has not yet yielded any clinically useful applications so far. The fate and contribution of the seeded cells are not sufficiently clarified, especially at clinically relevant locations. Therefore, we investigated cell proliferation around the spine and at ectopic sites using noninvasive in vivo bioluminescence imaging (BLI) in relation to new bone formation. Goat MSCs were lentivirally transduced to express luciferase. After showing both correlation between MSC viability and BLI signal as well as survival and osteogenic capacity of these cells ectopically in mice, they were seeded on ceramic scaffolds and implanted in immunodeficient rats at two levels in the spine for spinal fusion as well as subcutaneously. Nontransduced MSCs were used as a control group. All rats were monitored at day 1 and after that weekly until termination at week 7. In mice a BLI signal was observed during the whole observation period, indicating survival of the seeded MSCs, which was accompanied by osteogenic differentiation in vivo. However, these same MSCs showed a different response in the rat model, where the BLI signal was present until day 14, both in the spine and ectopically, indicating that MSCs were able to survive at least 2 weeks of implantation. Only when the signal was still present after the total implantation period ectopically, which only occurred in one rat, new bone was formed extensively and the implanted MSCs were responsible for this bone formation. Ectopically, neither a reduced proliferative group (irradiated) nor a group in which the cells were devitalized by liquid nitrogen and the produced extracellular matrix remained (matrix group) resulted in bone formation. This suggests that the release of soluble factors or the presence of an extracellular matrix is not enough to induce bone formation. For the spinal location, the question remains whether the implanted MSCs contribute to the bone regeneration or that the principal mechanism of MSC activity is through the release of soluble mediators.


Assuntos
Osso e Ossos/fisiologia , Luciferases/metabolismo , Células-Tronco Multipotentes/citologia , Fusão Vertebral/métodos , Coloração e Rotulagem , Engenharia Tecidual/métodos , Animais , Cabras , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Implantes Experimentais , Medições Luminescentes , Camundongos , Células-Tronco Multipotentes/metabolismo , Osteogênese/fisiologia , Ratos , Células Estromais/citologia , Alicerces Teciduais , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA