Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Radiology ; 310(2): e232365, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38349244

RESUMO

Background Image-guided tumor ablation is the first-line therapy for early-stage hepatocellular carcinoma (HCC), with ongoing investigations into its combination with immunotherapies. Matrix metalloproteinase (MMP) inhibition demonstrates immunomodulatory potential and reduces HCC tumor growth when combined with ablative treatment. Purpose To evaluate the effect of incomplete cryoablation with or without MMP inhibition on the local immune response in residual tumors in a murine HCC model. Materials and Methods Sixty 8- to 10-week-old female BALB/c mice underwent HCC induction with use of orthotopic implantation of syngeneic Tib-75 cells. After 7 days, mice with a single lesion were randomized into treatment groups: (a) no treatment, (b) MMP inhibitor, (c) incomplete cryoablation, and (d) incomplete cryoablation and MMP inhibitor. Macrophage and T-cell subsets were assessed in tissue samples with use of immunohistochemistry and immunofluorescence (cell averages calculated using five 1-µm2 fields of view [FOVs]). C-X-C motif chemokine receptor type 3 (CXCR3)- and interferon γ (IFNγ)-positive T cells were assessed using flow cytometry. Groups were compared using unpaired Student t tests, one-way analysis of variance with Tukey correction, and the Kruskal-Wallis test with Dunn correction. Results Mice treated with incomplete cryoablation (n = 6) showed greater infiltration of CD206+ tumor-associated macrophages (mean, 1.52 cells per FOV vs 0.64 cells per FOV; P = .03) and MMP9-expressing cells (mean, 0.89 cells per FOV vs 0.11 cells per FOV; P = .03) compared with untreated controls (n = 6). Incomplete cryoablation with MMP inhibition (n = 6) versus without (n = 6) led to greater CD8+ T-cell (mean, 15.8% vs 8.29%; P = .04), CXCR3+CD8+ T-cell (mean, 11.64% vs 8.47%; P = .004), and IFNγ+CD8+ T-cell infiltration (mean, 11.58% vs 5.18%; P = .02). Conclusion In a mouse model of HCC, incomplete cryoablation and systemic MMP inhibition showed increased cytotoxic CD8+ T-cell infiltration into the residual tumor compared with either treatment alone. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Gemmete in this issue.


Assuntos
Carcinoma Hepatocelular , Criocirurgia , Neoplasias Hepáticas , Feminino , Animais , Camundongos , Carcinoma Hepatocelular/cirurgia , Inibidores de Metaloproteinases de Matriz , Neoplasias Hepáticas/cirurgia , Linfócitos T CD8-Positivos , Metaloproteinases da Matriz
2.
Kidney Int ; 105(2): 218-230, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38245210

RESUMO

Glomerular diseases are classified using a descriptive taxonomy that is not reflective of the heterogeneous underlying molecular drivers. This limits not only diagnostic and therapeutic patient management, but also impacts clinical trials evaluating targeted interventions. The Nephrotic Syndrome Study Network (NEPTUNE) is poised to address these challenges. The study has enrolled >850 pediatric and adult patients with proteinuric glomerular diseases who have contributed to deep clinical, histologic, genetic, and molecular profiles linked to long-term outcomes. The NEPTUNE Knowledge Network, comprising combined, multiscalar data sets, captures each participant's molecular disease processes at the time of kidney biopsy. In this editorial, we describe the design and implementation of NEPTUNE Match, which bridges a basic science discovery pipeline with targeted clinical trials. Noninvasive biomarkers have been developed for real-time pathway analyses. A Molecular Nephrology Board reviews the pathway maps together with clinical, laboratory, and histopathologic data assembled for each patient to compile a Match report that estimates the fit between the specific molecular disease pathway(s) identified in an individual patient and proposed clinical trials. The NEPTUNE Match report is communicated using established protocols to the patient and the attending nephrologist for use in their selection of available clinical trials. NEPTUNE Match represents the first application of precision medicine in nephrology with the aim of developing targeted therapies and providing the right medication for each patient with primary glomerular disease.


Assuntos
Nefropatias , Síndrome Nefrótica , Adulto , Criança , Humanos , Biomarcadores , Ensaios Clínicos como Assunto , Glomérulos Renais/patologia , Síndrome Nefrótica/diagnóstico , Síndrome Nefrótica/genética , Síndrome Nefrótica/terapia
3.
Calcif Tissue Int ; 113(4): 437-448, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37566229

RESUMO

Quantification of in vitro osteoclast cultures (e.g. cell number) often relies on manual counting methods. These approaches are labour intensive, time consuming and result in substantial inter- and intra-user variability. This study aimed to develop and validate an automated workflow to robustly quantify in vitro osteoclast cultures. Using ilastik, a machine learning-based image analysis software, images of tartrate resistant acid phosphatase-stained mouse osteoclasts cultured on dentine discs were used to train the ilastik-based algorithm. Assessment of algorithm training showed that osteoclast numbers strongly correlated between manual- and automatically quantified values (r = 0.87). Osteoclasts were consistently faithfully segmented by the model when visually compared to the original reflective light images. The ability of this method to detect changes in osteoclast number in response to different treatments was validated using zoledronate, ticagrelor, and co-culture with MCF7 breast cancer cells. Manual and automated counting methods detected a 70% reduction (p < 0.05) in osteoclast number, when cultured with 10 nM zoledronate and a dose-dependent decrease with 1-10 µM ticagrelor (p < 0.05). Co-culture with MCF7 cells increased osteoclast number by ≥ 50% irrespective of quantification method. Overall, an automated image segmentation and analysis workflow, which consistently and sensitively identified in vitro osteoclasts, was developed. Advantages of this workflow are (1) significantly reduction in user variability of endpoint measurements (93%) and analysis time (80%); (2) detection of osteoclasts cultured on different substrates from different species; and (3) easy to use and freely available to use along with tutorial resources.


Assuntos
Reabsorção Óssea , Osteoclastos , Camundongos , Animais , Ácido Zoledrônico , Ticagrelor , Técnicas de Cocultura , Células Cultivadas , Fosfatase Ácida/análise , Fosfatase Ácida Resistente a Tartarato , Diferenciação Celular
4.
Bone ; 176: 116868, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37549801

RESUMO

Extracellular pyrophosphate (PPi) is well known for its fundamental role as a physiochemical mineralisation inhibitor. However, information about its direct actions on bone cells remains limited. This study shows that PPi decreased osteoclast formation and resorptive activity by ≤50 %. These inhibitory actions were associated with reduced expression of genes involved in osteoclastogenesis (Tnfrsf11a, Dcstamp) and bone resorption (Ctsk, Car2, Acp5). In osteoblasts, PPi present for the entire (0-21 days) or latter stages of culture (7-21/14-21 days) decreased bone mineralisation by ≤95 %. However, PPi present for the differentiation phase only (0-7/0-14 days) increased bone formation (≤70 %). Prolonged treatment with PPi resulted in earlier matrix deposition and increased soluble collagen levels (≤2.3-fold). Expression of osteoblast (RUNX2, Bglap) and early osteocyte (E11, Dmp1) genes along with mineralisation inhibitors (Spp1, Mgp) was increased by PPi (≤3-fold). PPi levels are regulated by tissue non-specific alkaline phosphatase (TNAP) and ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1). PPi reduced NPP1 expression in both cell types whereas TNAP expression (≤2.5-fold) and activity (≤35 %) were increased in osteoblasts. Breakdown of extracellular ATP by NPP1 represents a key source of PPi. ATP release from osteoclasts and osteoblasts was decreased ≤60 % by PPi and by a selective TNAP inhibitor (CAS496014-12-2). Pertussis toxin, which prevents Gαi subunit activation, was used to investigate whether G-protein coupled receptor (GPCR) signalling mediates the effects of PPi. The actions of PPi on bone mineralisation, collagen production, ATP release, gene/protein expression and osteoclast formation were abolished or attenuated by pertussis toxin. Together these findings show that PPi, modulates differentiation, function and gene expression in osteoblasts and osteoclasts. The ability of PPi to alter ATP release and NPP1/TNAP expression and activity indicates that cells can detect PPi levels and respond accordingly. Our data also raise the possibility that some actions of PPi on bone cells could be mediated by a Gαi-linked GPCR.


Assuntos
Difosfatos , Osteoclastos , Osteoclastos/metabolismo , Difosfatos/farmacologia , Toxina Pertussis/metabolismo , Toxina Pertussis/farmacologia , Osteoblastos/metabolismo , Colágeno/metabolismo , Trifosfato de Adenosina/metabolismo , Fosfatase Alcalina/metabolismo
5.
J Endocrinol ; 257(2)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36802398

RESUMO

As a key regulator of bone homeostasis, sclerostin has garnered a lot of interest over the last two decades. Although sclerostin is primarily expressed by osteocytes and is well known for its role in bone formation and remodelling, it is also expressed by a number of other cells and potentially plays a role in other organs. Herein, we aim to bring together recent sclerostin research and discuss the effect of sclerostin on bone, cartilage, muscle, liver, kidney and the cardiovascular and immune systems. Particular focus is placed on its role in diseases, such as osteoporosis and myeloma bone disease, and the novel development of sclerostin as a therapeutic target. Anti-sclerostin antibodies have recently been approved for the treatment of osteoporosis. However, a cardiovascular signal was observed, prompting extensive research into the role of sclerostin in vascular and bone tissue crosstalk. The study of sclerostin expression in chronic kidney disease was followed by the investigation of its role in liver-lipid-bone interactions, and the recent discovery of sclerostin as a myokine prompted new research into sclerostin within the bone-muscle relationship. Potentially, the effects of sclerostin reach beyond that of bone alone. We further summarise recent developments in the use of sclerostin as a potential therapeutic for osteoarthritis, osteosarcoma and sclerosteosis. Overall, these new treatments and discoveries illustrate progress within the field, however, also highlight remaining gaps in our knowledge.


Assuntos
Proteínas Morfogenéticas Ósseas , Osteoporose , Humanos , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Marcadores Genéticos , Osso e Ossos/metabolismo
6.
Cytotherapy ; 22(8): 424-435, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32522398

RESUMO

BACKGROUND: The periosteum is a highly vascularized, collagen-rich tissue that plays a crucial role in directing bone repair. This is orchestrated primarily by its resident progenitor cell population. Indeed, preservation of periosteum integrity is critical for bone healing. Cells extracted from the periosteum retain their osteochondrogenic properties and as such are a promising basis for tissue engineering strategies for the repair of bone defects. However, the culture expansion conditions and the way in which the cells are reintroduced to the defect site are critical aspects of successful translation. Indeed, expansion in human serum and implantation on biomimetic materials has previously been shown to improve in vivo bone formation. AIM: This study aimed to develop a protocol to allow for the expansion of human periosteum derived cells (hPDCs) in a biomimetic periosteal-like environment. METHODS: The expansion conditions were defined through the investigation of the bioactive cues involved in augmenting hPDC proliferative and multipotency characteristics, based on transcriptomic analysis of cells cultured in human serum. RESULTS: Master regulators of transcriptional networks were identified, and an optimized periosteum-derived growth factor cocktail (PD-GFC; containing ß-estradiol, FGF2, TNFα, TGFß, IGF-1 and PDGF-BB) was generated. Expansion of hPDCs in PD-GFC resulted in serum mimicry with regard to the cell morphology, proliferative capacity and chondrogenic differentiation. When incorporated into a three-dimensional collagen type 1 matrix and cultured in PD-GFC, the hPDCs migrated to the surface that represented the matrix topography of the periosteum cambium layer. Furthermore, gene expression analysis revealed a down-regulated WNT and TGFß signature and an up-regulation of CREB, which may indicate the hPDCs are recreating their progenitor cell signature. CONCLUSION: This study highlights the first stage in the development of a biomimetic periosteum, which may have applications in bone repair.


Assuntos
Materiais Biomiméticos/farmacologia , Redes Reguladoras de Genes , Periósteo/patologia , Soro/metabolismo , Adolescente , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Condrogênese/efeitos dos fármacos , Colágeno Tipo I/farmacologia , Feminino , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Periósteo/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
7.
Sci Rep ; 8(1): 2942, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29440666

RESUMO

Neural tube defects (NTDs) are severe congenital abnormalities, caused by failed closure of neural tube during early embryonic development. Periconceptional folic acid (FA) supplementation greatly reduces the risk of NTDs. However, the molecular mechanisms behind NTDs and the preventive role of FA remain unclear. Here, we use human induced pluripotent stem cells (iPSCs) derived from fetuses with spina bifida aperta (SBA) to study the pathophysiology of NTDs and explore the effects of FA exposure. We report that FA exposure in SBA model is necessary for the proper formation and maturation of neural tube structures and robust differentiation of mesodermal derivatives. Additionally, we show that the folate antagonist methotrexate dramatically affects the formation of neural tube structures and FA partially reverts this aberrant phenotype. In conclusion, we present a novel model for human NTDs and provide evidence that it is a powerful tool to investigate the molecular mechanisms underlying NTDs, test drugs for therapeutic approaches.


Assuntos
Ácido Fólico/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Fenótipo , Espinha Bífida Cística/patologia , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX7/genética , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
8.
Tissue Eng Part C Methods ; 22(5): 473-86, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27018617

RESUMO

Successful application of cell-based strategies in cartilage and bone tissue engineering has been hampered by the lack of robust protocols to efficiently differentiate mesenchymal stem cells into the chondrogenic lineage. The development of chemically defined culture media supplemented with growth factors (GFs) has been proposed as a way to overcome this limitation. In this work, we applied a fractional design of experiment (DoE) strategy to screen the effect of multiple GFs (BMP2, BMP6, GDF5, TGF-ß1, and FGF2) on chondrogenic differentiation of human periosteum-derived mesenchymal stem cells (hPDCs) in vitro. In a micromass culture (µMass) system, BMP2 had a positive effect on glycosaminoglycan deposition at day 7 (p < 0.001), which in combination with BMP6 synergistically enhanced cartilage-like tissue formation that displayed in vitro mineralization capacity at day 14 (p < 0.001). Gene expression of µMasses cultured for 7 days with a medium formulation supplemented with 100 ng/mL of BMP2 and BMP6 and a low concentration of GDF5, TGF-ß1, and FGF2 showed increased expression of Sox9 (1.7-fold) and the matrix molecules aggrecan (7-fold increase) and COL2A1 (40-fold increase) compared to nonstimulated control µMasses. The DoE analysis indicated that in GF combinations, BMP2 was the strongest effector for chondrogenic differentiation of hPDCs. When transplanted ectopically in nude mice, the in vitro-differentiated µMasses showed maintenance of the cartilaginous phenotype after 4 weeks in vivo. This study indicates the power of using the DoE approach for the creation of new medium formulations for skeletal tissue engineering approaches.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Condrócitos/citologia , Condrogênese/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Periósteo/citologia , Engenharia Tecidual/métodos , Adolescente , Animais , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Periósteo/efeitos dos fármacos , Periósteo/metabolismo , Doadores de Tecidos
9.
Biomaterials ; 86: 106-18, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26901484

RESUMO

The development of osteoinductive calcium phosphate- (CaP) based biomaterials has, and continues to be, a major focus in the field of bone tissue engineering. However, limited insight into the spatiotemporal activation of signalling pathways has hampered the optimisation of in vivo bone formation and subsequent clinical translation. To gain further knowledge regarding the early molecular events governing bone tissue formation, we combined human periosteum derived progenitor cells with three types of clinically used CaP-scaffolds, to obtain constructs with a distinct range of bone forming capacity in vivo. Protein phosphorylation together with gene expression for key ligands and target genes were investigated 24 hours after cell seeding in vitro, and 3 and 12 days post ectopic implantation in nude mice. A computational modelling approach was used to deduce critical factors for bone formation 8 weeks post implantation. The combined Ca(2+)-mediated activation of BMP-, Wnt- and PKC signalling pathways 3 days post implantation were able to discriminate the bone forming from the non-bone forming constructs. Subsequently, a mathematical model able to predict in vivo bone formation with 96% accuracy was developed. This study illustrates the importance of defining and understanding CaP-activated signalling pathways that are required and sufficient for in vivo bone formation. Furthermore, we demonstrate the reliability of mathematical modelling as a tool to analyse and deduce key factors within an empirical data set and highlight its relevance to the translation of regenerative medicine strategies.


Assuntos
Materiais Biocompatíveis/química , Fosfatos de Cálcio/química , Osteogênese , Transdução de Sinais , Células-Tronco/citologia , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/metabolismo , Materiais Biocompatíveis/farmacologia , Cálcio/metabolismo , Fosfatos de Cálcio/metabolismo , Fosfatos de Cálcio/farmacologia , Células Cultivadas , Humanos , Camundongos Nus , Osteogênese/efeitos dos fármacos , Periósteo/citologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Engenharia Tecidual , Via de Sinalização Wnt/efeitos dos fármacos
10.
PLoS One ; 9(4): e94833, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24747914

RESUMO

PURPOSE: The use of stably integrated reporter gene imaging provides a manner to monitor the in vivo fate of engrafted cells over time in a non-invasive manner. Here, we optimized multimodal imaging (small-animal PET, Cerenkov luminescence imaging (CLI) and bioluminescence imaging (BLI)) of mesenchymal stem cells (MSCs), by means of the human sodium iodide symporter (hNIS) and firefly luciferase (Fluc) as reporters. METHODS: First, two multicistronic lentiviral vectors (LV) were generated for multimodal imaging: BLI, 124I PET/SPECT and CLI. Expression of the imaging reporter genes was validated in vitro using 99mTcO4- radioligand uptake experiments and BLI. Uptake kinetics, specificity and tracer elution were determined as well as the effect of the transduction process on the cell's differentiation capacity. MSCs expressing the LV were injected intravenously or subcutaneously and imaged using small-animal PET, CLI and BLI. RESULTS: The expression of both imaging reporter genes was functional and specific. An elution of 99mTcO4- from the cells was observed, with 31% retention after 3 h. After labeling cells with 124I in vitro, a significantly higher CLI signal was noted in hNIS expressing murine MSCs. Furthermore, it was possible to visualize cells injected intravenously using BLI or subcutaneously in mice, using 124I small-animal PET, CLI and BLI. CONCLUSIONS: This study identifies hNIS as a suitable reporter gene for molecular imaging with PET and CLI, as confirmed with BLI through the expression of Fluc. It supports the potential for a wider application of hNIS reporter gene imaging and future clinical applications.


Assuntos
Luminescência , Células-Tronco Mesenquimais/metabolismo , Imagem Multimodal/métodos , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Simportadores/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Genes Reporter/genética , Vetores Genéticos/genética , Células HEK293 , Humanos , Radioisótopos do Iodo , Lentivirus/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Puromicina/farmacologia
11.
Cell Reprogram ; 16(1): 29-39, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24459991

RESUMO

Strategies for bone regeneration are undergoing a paradigm shift, moving away from the replication of end-stage bone tissue and instead aiming to recapture the initial events of fracture repair. Although this is known to resemble endochondral bone formation, chondrogenic cell types with favorable proliferative and hypertrophic differentiation properties are lacking. Recent advances in cellular reprogramming have allowed the creation of alternative cell populations with specific properties through the forced expression of transcription factors. Herein, we investigated the in vitro hypertrophic differentiation and in vivo tissue formation capacity of induced chondrogenic cells (iChon cells) obtained through direct reprogramming. In vitro hypertrophic differentiation was detected in iChon cells that contained a doxycycline-inducible expression system for Klf4, cMyc, and Sox9. Furthermore, endochondral bone formation was detected after implantation in nude mice. The bone tissue was derived entirely from host origin, whereas cartilage tissue contained cells from both host and donor. The results obtained highlight the promise of cellular reprogramming for the creation of functional skeletal cells that can be used for novel bone healing strategies.


Assuntos
Diferenciação Celular , Derme/metabolismo , Fibroblastos/metabolismo , Osteogênese , Fatores de Transcrição SOX9/metabolismo , Animais , Linhagem Celular , Derme/citologia , Fibroblastos/citologia , Fibroblastos/transplante , Xenoenxertos , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOX9/genética
12.
Tissue Eng Part A ; 19(15-16): 1817-28, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23530894

RESUMO

Human mesenchymal stromal cells (hMSCs) offer great potential for bone tissue engineering applications, but their in vivo performance remains limited. Preconditioning of these cells with small molecules to improve their differentiation before implantation, or incorporation of growth factors are possible solutions. Insulin-like growth factor-1 (IGF-1) is one of the most abundant growth factors in bone, involved in growth, development, and metabolism, but its effects on hMSCs are still subject of debate. Here we examined the effects of IGF-1 on proliferation and differentiation of hMSCs in vitro and we found that serum abolished the effects of IGF-1. Only in the absence of serum, IGF-1 increased proliferation, alkaline phosphatase expression, and osteogenic gene expression of hMSCs. Furthermore, we examined synergistic effects of bone morphogenetic protein-2 (BMP-2) and IGF-1 and, although IGF-1 enhanced BMP-2-induced mineralization, IGF-1 only slightly affected in vivo bone formation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletroquímica de Varredura
13.
J Nucl Med ; 54(3): 447-54, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23353687

RESUMO

UNLABELLED: Because of their extended differentiation capacity, stem cells have gained great interest in the field of regenerative medicine. For the development of therapeutic strategies, more knowledge on the in vivo fate of these cells has to be acquired. Therefore, stem cells can be labeled with radioactive tracer molecules such as (18)F-FDG, a positron-emitting glucose analog that is taken up and metabolically trapped by the cells. The aim of this study was to optimize the radioactive labeling of mesenchymal stem cells (MSCs) and multipotent adult progenitor cells (MAPCs) in vitro with (18)F-FDG and to investigate the potential radiotoxic effects of this labeling procedure with a range of techniques, including transmission electron microscopy (TEM). METHODS: Mouse MSCs and rat MAPCs were used for (18)F-FDG uptake kinetics and tracer retention studies. Cell metabolic activity, proliferation, differentiation and ultrastructural changes after labeling were evaluated using an Alamar Blue reagent, doubling time calculations and quantitative TEM, respectively. Additionally, mice were injected with MSCs and MAPCs prelabeled with (18)F-FDG, and stem cell biodistribution was investigated using small-animal PET. RESULTS: The optimal incubation period for (18)F-FDG uptake was 60 min. Significant early tracer washout was observed, with approximately 30%-40% of the tracer being retained inside the cells 3 h after labeling. Cell viability, proliferation, and differentiation capacity were not severely affected by (18)F-FDG labeling. No major changes at the ultrastructural level, considering mitochondrial length, lysosome size, the number of lysosomes, the number of vacuoles, and the average rough endoplasmic reticulum width, were observed with TEM. Small-animal PET experiments with radiolabeled MAPCs and MSCs injected intravenously in mice showed a predominant accumulation in the lungs and a substantial elution of (18)F-FDG from the cells. CONCLUSION: MSCs and MAPCs can be successfully labeled with (18)F-FDG for molecular imaging purposes. The main cellular properties are not rigorously affected. TEM confirmed that the cells' ultrastructural properties are not influenced by (18)F-FDG labeling. Small-animal PET studies confirmed the intracellular location of the tracer and the possibility of imaging injected prelabeled stem cell types in vivo. Therefore, direct labeling of MSCs and MAPCs with (18)F-FDG is a suitable technique to noninvasively assess cell delivery and early retention with PET.


Assuntos
Células-Tronco Adultas/diagnóstico por imagem , Fluordesoxiglucose F18 , Células-Tronco Mesenquimais/diagnóstico por imagem , Células-Tronco Multipotentes/diagnóstico por imagem , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/ultraestrutura , Animais , Diferenciação Celular , Células Cultivadas , Radioisótopos de Flúor , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Microscopia Eletrônica de Transmissão , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/ultraestrutura , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Ratos , Medicina Regenerativa , Engenharia Tecidual
14.
Stem Cells ; 30(11): 2460-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22911908

RESUMO

One of the key challenges in bone tissue engineering is the timely formation of blood vessels that promote the survival of the implanted cells in the construct. Fracture healing largely depends on the presence of an intact periosteum but it is still unknown whether periosteum-derived cells (PDC) are critical for bone repair only by promoting bone formation or also by inducing neovascularization. We first established a protocol to specifically isolate murine PDC (mPDC) from long bones of adult mice. Mesenchymal stem cells were abundantly present in this cell population as more than 50% of the mPDC expressed mesenchymal markers (CD73, CD90, CD105, and stem cell antigen-1) and the cells exhibited trilineage differentiation potential (chondrogenic, osteogenic, and adipogenic). When transplanted on a collagen-calcium phosphate scaffold in vivo, mPDC attracted numerous blood vessels and formed mature bone which comprises a hematopoiesis-supportive stroma. We explored the proangiogenic properties of mPDC using in vitro culture systems and showed that mPDC promote the survival and proliferation of endothelial cells through the production of vascular endothelial growth factor. Coimplantation with endothelial cells demonstrated that mPDC can enhance vasculogenesis by adapting a pericyte-like phenotype, in addition to their ability to stimulate blood vessel ingrowth from the host. In conclusion, these findings demonstrate that periosteal cells contribute to fracture repair, not only through their strong osteogenic potential but also through their proangiogenic features and thus provide an ideal cell source for bone regeneration therapies.


Assuntos
Osso e Ossos/irrigação sanguínea , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Osteogênese , Periósteo/citologia , Animais , Antígenos CD/metabolismo , Regeneração Óssea , Substitutos Ósseos , Osso e Ossos/citologia , Osso e Ossos/fisiologia , Fosfatos de Cálcio , Diferenciação Celular , Hipóxia Celular , Separação Celular , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Colágeno , Feminino , Citometria de Fluxo , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Cultura Primária de Células , Engenharia Tecidual , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Biomaterials ; 33(16): 4044-58, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22381474

RESUMO

Successful clinical repair of non-healing skeletal defects requires the use of bone substitutes with robust bone inductivity and excellent biomechanical stability. Thus, three-dimensionally functionalised porous calcium phosphate-Ti6Al4V (CaP-Ti) hybrids were produced by perfusion electrodeposition, and the in vitro and in vivo biological performances were evaluated using human periosteum derived cells (hPDCs). By applying various current densities at the optimised deposition conditions, CaP coatings with sub-micrometer to nano-scale porous crystalline structures and different ion dissolution kinetics were deposited on the porous Ti6Al4V scaffolds. These distinctive physicochemical properties caused a significant impact on in vitro proliferation, osteogenic differentiation, and matrix mineralisation of hPDCs. This includes a potential role of hPDCs in mediating osteoclastogenesis for the resorption of CaP coatings, as indicated by a significant down-regulation of osteoprotegerin (OPG) gene expression and by the histological observation of abundant multi-nucleated giant cells near to the coatings. By subcutaneous implantation, the produced hybrids induced ectopic bone formation, which was highly dependent on the physicochemical properties of the CaP coating (including the Ca(2+) dissolution kinetics and coating surface topography), in a cell density-dependent manner. This study provided further insight on stem cell-CaP biomaterial interactions, and the feasibility to produced bone reparative units that are predictively osteoinductive in vivo by perfusion electrodeposition technology.


Assuntos
Desenvolvimento Ósseo , Fosfatos de Cálcio/química , Técnicas Eletroquímicas , Titânio/química , Ligas , Sequência de Bases , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Primers do DNA , Regulação para Baixo , Humanos , Microscopia Eletrônica de Varredura , Osteoprotegerina/genética , Reação em Cadeia da Polimerase , Solubilidade , Propriedades de Superfície , Tomografia Computadorizada por Raios X
16.
Science ; 335(6064): 104-8, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22223807

RESUMO

Polyaromatic hydrocarbons (PAHs) are prevalent, potent carcinogens, and 7,12-dimethylbenz[a]anthracene (DMBA) is a model PAH widely used to study tumorigenesis. Mice lacking Langerhans cells (LCs), a signatory epidermal dendritic cell (DC), are protected from cutaneous chemical carcinogenesis, independent of T cell immunity. Investigation of the underlying mechanism revealed that LC-deficient skin was relatively resistant to DMBA-induced DNA damage. LCs efficiently metabolized DMBA to DMBA-trans-3,4-diol, an intermediate proximal to oncogenic Hras mutation, and DMBA-treated LC-deficient skin contained significantly fewer Hras mutations. Moreover, DMBA-trans-3,4-diol application bypassed tumor resistance in LC-deficient mice. Additionally, the genotoxic impact of DMBA on human keratinocytes was significantly increased by prior incubation with human-derived LC. Thus, tissue-associated DC can enhance chemical carcinogenesis via PAH metabolism, highlighting the complex relation between immune cells and carcinogenesis.


Assuntos
9,10-Dimetil-1,2-benzantraceno/análogos & derivados , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/induzido quimicamente , Dano ao DNA , Células de Langerhans/metabolismo , Neoplasias Cutâneas/induzido quimicamente , 9,10-Dimetil-1,2-benzantraceno/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Carcinoma de Células Escamosas/metabolismo , Transformação Celular Neoplásica , Células Cultivadas , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1B1 , Genes ras , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Células de Langerhans/imunologia , Camundongos , Camundongos Transgênicos , Neoplasias Cutâneas/metabolismo , Linfócitos T/imunologia
17.
Stem Cells ; 29(5): 871-82, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21433224

RESUMO

Several adherent postnatal stem cells have been described with different phenotypic and functional properties. As many of these cells are being considered for clinical therapies, it is of great importance that the identity and potency of these products is validated. We compared the phenotype and functional characteristics of human mesenchymal stem cells (hMSCs), human mesoangioblasts (hMab), and human multipotent adult progenitor cells (hMAPCs) using uniform standardized methods. Human MAPCs could be expanded significantly longer in culture. Differences in cell surface marker expression were found among the three cell populations with CD140b being a distinctive marker among the three cell types. Differentiation capacity towards adipocytes, osteoblasts, chondrocytes, and smooth muscle cells in vitro, using established protocols, was similar among the three cell types. However, only hMab differentiated to skeletal myocytes, while only hMAPCs differentiated to endothelium in vitro and in vivo. A comparative transcriptome analysis confirmed that the three cell populations are distinct and revealed gene signatures that correlated with their specific functional properties. Furthermore, we assessed whether the phenotypic, functional, and transcriptome features were mediated by the culture conditions. Human MSCs and hMab cultured under MAPC conditions became capable of generating endothelial-like cells, whereas hMab lost some of their ability to generate myotubes. By contrast, hMAPCs cultured under MSC conditions lost their endothelial differentiation capacity, whereas this was retained when cultured under Mab conditions, however, myogenic capacity was not gained under Mab conditions. These studies demonstrate that hMSCs, hMab, and hMAPCs have different properties that are partially mediated by the culture conditions.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Adipócitos/citologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Condrócitos/citologia , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Miócitos de Músculo Liso/citologia , Osteoblastos/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Tissue Eng Part A ; 17(7-8): 1083-97, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21091326

RESUMO

The use of calcium phosphate (CaP)-based carriers in bone engineering is a promising approach to induce in vivo bone formation. However, the exact mechanism of osteoinduction by CaP is not known. Here, by mimicking the in vivo Ca(2+) and P(i)-enriched environment in an in vitro model, we assessed the effects of these ions on human periosteum-derived cells. We observed a significant Ca(2+) and P(i)-induced cell proliferation, which was found to be through the modulation of cell cycle progression, in a dose- and time-dependent manner. In addition, Ca(2+), P(i), and combined Ca-P upregulated osteogenic gene expression in a dose- and time-dependent manner. Encouragingly, both ions administered individually or in combination persistently and dose dependently upregulated bone morphogenetic protein-2 gene expression. This suggested a potential osteoinductive effect through an autonomous activation of the bone morphogenetic protein signaling pathway by released Ca(2+) and P(i), which may serve as an autocrine/paracrine osteoinduction loop that drives the cellularized CaP constructs toward effective bone formation in vivo. In conclusion, through an in vitro biomimetic model, we have partially probed the roles of the released Ca(2+) and P(i) on the osteoinductivity of CaP-based biomaterials.


Assuntos
Materiais Biocompatíveis/química , Fosfatos de Cálcio/farmacologia , Proteína Morfogenética Óssea 2/genética , Fosfatos de Cálcio/química , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Osteocalcina/genética , Osteopontina/genética , Periósteo/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
J Invest Dermatol ; 130(6): 1726-36, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19924136

RESUMO

T-pro are tumor-infiltrating TCRalphabeta(+)CD8(+) cells of reduced cytotoxic potential that promote experimental two-stage chemical cutaneous carcinogenesis. Toward understanding their mechanism of action, this study uses whole-genome expression analysis to compare T-pro with systemic CD8(+) T cells from multiple groups of tumor-bearing mice. T-pro show an overt T helper 17-like profile (high retinoic acid-related orphan receptor-(ROR)gammat, IL-17A, IL-17F; low T-bet and eomesodermin), regulatory potential (high FoxP3, IL-10, Tim-3), and transcripts encoding epithelial growth factors (amphiregulin, Gro-1, Gro-2). Tricolor flow cytometry subsequently confirmed the presence of TCRbeta(+) CD8(+) IL-17(+) T cells among tumor-infiltrating lymphocytes (TILs). Moreover, a time-course analysis of independent TIL isolates from papillomas versus carcinomas exposed a clear association of the "T-pro phenotype" with malignant progression. This molecular characterization of T-pro builds a foundation for elucidating the contributions of inflammation to cutaneous carcinogenesis, and may provide useful biomarkers for cancer immunotherapy in which the widely advocated use of tumor-specific CD8(+) cytolytic T cells should perhaps accommodate the cells' potential corruption toward the T-pro phenotype. The data are also likely germane to psoriasis, in which the epidermis may be infiltrated by CD8(+) IL-17-producing T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Perfilação da Expressão Gênica , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , 9,10-Dimetil-1,2-benzantraceno/efeitos adversos , Anfirregulina , Animais , Diferenciação Celular , Modelos Animais de Doenças , Família de Proteínas EGF , Fatores de Transcrição Forkhead/metabolismo , Glicoproteínas/metabolismo , Receptor Celular 2 do Vírus da Hepatite A , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores Virais/metabolismo , Neoplasias Cutâneas/induzido quimicamente
20.
Nat Immunol ; 10(4): 427-36, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270712

RESUMO

The production of cytokines such as interferon-gamma and interleukin 17 by alphabeta and gammadelta T cells influences the outcome of immune responses. Here we show that most gammadelta T lymphocytes expressed the tumor necrosis factor receptor family member CD27 and secreted interferon-gamma, whereas interleukin 17 production was restricted to CD27(-) gammadelta T cells. In contrast to the apparent plasticity of alphabeta T cells, the cytokine profiles of these distinct gammadelta T cell subsets were essentially stable, even during infection. These phenotypes were established during thymic development, when CD27 functions as a regulator of the differentiation of gammadelta T cells at least in part by inducing expression of the lymphotoxin-beta receptor and genes associated with trans-conditioning and interferon-gamma production. Thus, the cytokine profiles of peripheral gammadelta T cells are predetermined mainly by a mechanism involving CD27.


Assuntos
Interferon gama/imunologia , Interleucina-17/imunologia , Células Progenitoras Linfoides/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Subpopulações de Linfócitos T/imunologia , Timo/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Ligante CD27/imunologia , Células Cultivadas , Receptor beta de Linfotoxina/imunologia , Malária Cerebral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA