Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Acta Biomater ; 149: 111-125, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35835287

RESUMO

Rapid vascularization of clinical-size bone grafts is an unsolved challenge in regenerative medicine. Vascular endothelial growth factor-A (VEGF) is the master regulator of angiogenesis. Its over-expression by genetically modified human osteoprogenitors has been previously evaluated to drive vascularization in osteogenic grafts, but has been observed to cause paradoxical bone loss through excessive osteoclast recruitment. However, during bone development angiogenesis and osteogenesis are physiologically coupled by VEGF expression. Here we investigated whether the mode of VEGF delivery may be a key to recapitulate its physiological function. VEGF activity requires binding to the extracellular matrix, and heterogeneous levels of expression lead to localized microenvironments of excessive dose. Therefore we hypothesized that a homogeneous distribution of matrix-associated factor in the microenvironment may enable efficient coupling of angiogenesis and bone formation. This was achieved by decorating fibrin matrices with a cross-linkable engineered version of VEGF (TG-VEGF) that is released only by enzymatic cleavage by invading cells. In ectopic grafts, both TG-VEGF and VEGF-expressing progenitors similarly improved vascularization within the first week, but efficient bone formation was possible only in the factor-decorated matrices, whereas heterogenous, cell-based VEGF expression caused significant bone loss. In critical-size orthotopic calvaria defects, TG-VEGF effectively improved early vascular invasion, osteoprogenitor survival and differentiation, as well as bone repair compared to both controls and VEGF-expressing progenitors. In conclusion, homogenous distribution of matrix-associated VEGF protein preserves the physiological coupling of angiogenesis and osteogenesis, providing an attractive and clinically applicable strategy to engineer vascularized bone. STATEMENT OF SIGNIFICANCE: The therapeutic regeneration of vascularized bone is an unsolved challenge in regenerative medicine. Stimulation of blood vessel growth by over-expression of VEGF has been associated with paradoxical bone loss, whereas angiogenesis and osteogenesis are physiologically coupled by VEGF during development. Here we found that controlling the distribution of VEGF dose in an osteogenic graft is key to recapitulate its physiological function. In fact, homogeneous decoration of fibrin matrices with engineered VEGF could improve both vascularization and bone formation in orthotopic critical-size defects, dispensing with the need for combined osteogenic factor delivery. VEGF-decorated fibrin matrices provide a readily translatable platform for engineering a controlled microenvironment for bone regeneration.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Regeneração Óssea , Fibrina/metabolismo , Fibrina/farmacologia , Humanos , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
2.
Cell Rep ; 32(10): 108105, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32905777

RESUMO

Circulating tumor cells (CTCs) are shed from solid cancers in the form of single or clustered cells, and the latter display an extraordinary ability to initiate metastasis. Yet, the biological phenomena that trigger the shedding of CTC clusters from a primary cancerous lesion are poorly understood. Here, when dynamically labeling breast cancer cells along cancer progression, we observe that the majority of CTC clusters are undergoing hypoxia, while single CTCs are largely normoxic. Strikingly, we find that vascular endothelial growth factor (VEGF) targeting leads to primary tumor shrinkage, but it increases intra-tumor hypoxia, resulting in a higher CTC cluster shedding rate and metastasis formation. Conversely, pro-angiogenic treatment increases primary tumor size, yet it dramatically suppresses the formation of CTC clusters and metastasis. Thus, intra-tumor hypoxia leads to the formation of clustered CTCs with high metastatic ability, and a pro-angiogenic therapy suppresses metastasis formation through prevention of CTC cluster generation.


Assuntos
Hipóxia Celular/imunologia , Células Neoplásicas Circulantes/imunologia , Proteômica/métodos , Animais , Feminino , Humanos , Masculino , Camundongos
3.
J Tissue Eng Regen Med ; 14(10): 1513-1523, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32841501

RESUMO

Therapeutic angiogenesis is the delivery of factors to promote vascular growth and holds promise for the treatment of ischemic heart conditions. Recombinant protein delivery to the myocardium by factor-decorated fibrin matrices is an attractive approach, thanks to the ability to precisely control both dose and duration of the treatment, the use of a clinically approved material like fibrin, and the avoidance of genetic modification. Here, we investigated the feasibility of inducing therapeutic angiogenesis in the rat myocardium by a state-of-the-art fibrin-based delivery platform that we previously optimized. Engineered versions of murine vascular endothelial growth factor A (VEGF164 ) and platelet-derived growth factor BB (PDGF-BB) were fused with an octapeptide substrate of the transglutaminase coagulation factor fXIIIa (TG) to allow their covalent cross-linking into fibrin hydrogels and release by enzymatic cleavage. Hydrogels containing either 100 µg/mL TG-VEGF alone or in combination with 10 µg/mL TG-PDGF-BB or no factor were injected into rat myocardium. Surprisingly, vascular density was severely reduced in all conditions, both in and around the injection site, where large fibrotic scars were formed. Scar formation was not due to the presence of growth factors, adaptive immunity to human proteins, damage from injection, nor to mechanical trauma from the hydrogel stiffness or volume. Rather scar was induced directly by fibrin and persisted despite hydrogel degradation within 1 week. These results caution against the suitability of fibrin-based platforms for myocardial growth factor delivery, despite their efficacy in other tissues, like skeletal muscle. The underlying molecular mechanisms must be further investigated in order to identify rational targets to prevent this serious side effect.


Assuntos
Cicatriz/patologia , Fibrina/efeitos adversos , Coração/efeitos dos fármacos , Hidrogéis/efeitos adversos , Neovascularização Fisiológica , Imunidade Adaptativa , Indutores da Angiogênese/metabolismo , Animais , Fenômenos Biomecânicos , Humanos , Injeções , Infarto do Miocárdio/patologia , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Stem Cells Transl Med ; 9(4): 433-444, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31922362

RESUMO

Therapeutic angiogenesis, that is, the generation of new vessels by delivery of specific factors, is required both for rapid vascularization of tissue-engineered constructs and to treat ischemic conditions. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis. However, uncontrolled expression can lead to aberrant vascular growth and vascular tumors (angiomas). Major challenges to fully exploit VEGF potency for therapy include the need to precisely control in vivo distribution of growth factor dose and duration of expression. In fact, the therapeutic window of VEGF delivery depends on its amount in the microenvironment around each producing cell rather than on the total dose, since VEGF remains tightly bound to extracellular matrix (ECM). On the other hand, short-term expression of less than about 4 weeks leads to unstable vessels, which promptly regress following cessation of the angiogenic stimulus. Here, we will briefly overview some key aspects of the biology of VEGF and angiogenesis and discuss their therapeutic implications with a particular focus on approaches using gene therapy, genetically modified progenitors, and ECM engineering with recombinant factors. Lastly, we will present recent insights into the mechanisms that regulate vessel stabilization and the switch between normal and aberrant vascular growth after VEGF delivery, to identify novel molecular targets that may improve both safety and efficacy of therapeutic angiogenesis.


Assuntos
Neovascularização Fisiológica , Medicina Regenerativa , Animais , Técnicas de Transferência de Genes , Humanos , Monócitos/citologia , Pericitos/citologia , Engenharia de Proteínas
5.
Swiss Med Wkly ; 149: w20011, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30685867

RESUMO

Despite major advances in medical, catheter-based or surgical treatment, cardiovascular diseases such as peripheral artery disease and coronary artery disease still cause significant morbidity and mortality. Furthermore, many patients do not qualify for catheter-based treatment or bypass surgery because of advanced disease or surgical risk. There is therefore an urgent need for novel treatment strategies. Therapeutic angiogenesis aims to restore blood flow to ischaemic tissue by stimulating the growth of new blood vessels through the local delivery of angiogenic factors, and may thus be an attractive treatment alternative for these patients. Angiogenesis is a complex process and the growth of normal, stable and functional vasculature depends on the coordinated interplay of different cell types and growth factors. Vascular endothelial growth factor-A (VEGF) is the fundamental regulator of vascular growth and the key target of therapeutic angiogenesis approaches. However, first-generation clinical trials of VEGF gene therapy have been disappointing, and a clear clinical benefit has yet to be established. In particular, VEGF delivery (a) appears to have a very limited therapeutic window in vivo: low doses are safe but mostly inefficient, whereas higher doses become rapidly unsafe; and (b) requires a sustained expression in vivo of at least about four weeks to achieve stable vessels that persist after cessation of the angiogenic stimulus. Here we will review the current understanding of how VEGF induces the growth of normal or pathological blood vessels, what limitations for the controlled induction of safe and efficient angiogenesis are intrinsically linked to the biological properties of VEGF, and how this knowledge can guide the design of more effective strategies for therapeutic angiogenesis.


Assuntos
Isquemia/terapia , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Indutores da Angiogênese/administração & dosagem , Animais , Becaplermina/administração & dosagem , Humanos
6.
Biotechnol J ; 12(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28881093

RESUMO

Bone marrow-derived mesenchymal stromal cells (BMSC), when expanded directly within 3D ceramic scaffolds in perfusion bioreactors, more reproducibly form bone when implanted in vivo as compared to conventional expansion on 2D polystyrene dishes/flasks. Since the bioreactor-based expansion on 3D ceramic scaffolds encompasses multiple aspects that are inherently different from expansion on 2D polystyrene, we aimed to decouple the effects of specific parameters among these two model systems. We assessed the effects of the: 1) 3D scaffold vs. 2D surface; 2) ceramic vs. polystyrene materials; and 3) BMSC niche established within the ceramic pores during in vitro culture, on subsequent in vivo bone formation. While BMSC expanded on 3D polystyrene scaffolds in the bioreactor could maintain their in vivo osteogenic potential, results were similar as BMSC expanded in monolayer on 2D polystyrene, suggesting little influence of the scaffold 3D environment. Bone formation was most reproducible when BMSC are expanded on 3D ceramic, highlighting the influence of the ceramic substrate. The presence of a pre-formed niche within the scaffold pores had negligible effects on the in vivo bone formation. The results of this study allow a greater understanding of the parameters required for perfusion bioreactor-based manufacturing of osteogenic grafts for clinical applications.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/instrumentação , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Alicerces Teciduais , Adolescente , Adulto , Reatores Biológicos , Técnicas de Cultura de Células/métodos , Proliferação de Células , Cerâmica/química , Humanos , Pessoa de Meia-Idade , Perfusão , Adulto Jovem
7.
J Tissue Eng Regen Med ; 11(10): 2836-2845, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-27296669

RESUMO

The present study reports on the development of an innovative culture substrate, micro-fabricated by two-photon laser polymerization (2PP) in a hybrid organic-inorganic photoresin. It was previously demonstrated that this substrate is able to guide spontaneous homing and colonization of mesenchymal stromal cells by the presence of synthetic microniches. Here, the number of niches covering the culture substrate was increased up to 10% of the total surface. Human bone marrow-derived mesenchymal stromal cells were expanded for 3 weeks and then their proliferation, clonogenic capacity and bilineage differentiation potential towards the osteogenic and adipogenic lineage were evaluated, both by colorimetric assays and by real-time polymerase chain reaction. Compared with cells cultured on glass substrates, cells expanded on 2PP substrates showed a greater colony diameter, which is an index of clonogenic potential. Following medium conditioning on 2PP-cultured cells, the expression of RUNX2 and BSP genes, as well as PPAR-gamma, was significantly greater than that measured on glass controls. Thus, human cells expanded on the synthetic niche substrate maintained their proliferative potential, clonogenic capacity and bilineage differentiation potential more effectively than cells expanded on glass substrates and in some aspects were comparable to non-expanded cells. © 2016 The Authors Journal of Tissue Engineering and Regenerative Medicine Published by John Wiley & Sons Ltd.


Assuntos
Lasers , Células-Tronco Mesenquimais/citologia , Fótons , Polimerização , Engenharia Tecidual/métodos , Adipogenia , Adolescente , Adulto , Idoso , Contagem de Células , Linhagem da Célula , Proliferação de Células , Células Clonais , Humanos , Pessoa de Meia-Idade , Osteogênese , Adulto Jovem
8.
Stem Cells Transl Med ; 5(8): 1090-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27334490

RESUMO

UNLABELLED: : Recapitulation of endochondral ossification (ECO) (i.e., generation of marrow-containing ossicles through a cartilage intermediate) has relevance to develop human organotypic models for bone or hematopoietic cells and to engineer grafts for bone regeneration. Unlike bone marrow-derived stromal cells (also known as bone marrow-derived mesenchymal stromal/stem cells), adipose-derived stromal cells (ASC) have so far failed to form a bone organ by ECO. The goal of the present study was to assess whether priming human ASC to a defined stage of chondrogenesis in vitro allows their autonomous ECO upon ectopic implantation. ASC were cultured either as micromass pellets or into collagen sponges in chondrogenic medium containing transforming growth factor-ß3 and bone morphogenetic protein-6 for 4 weeks (early hypertrophic templates) or for two additional weeks in medium supplemented with ß-glycerophosphate, l-thyroxin, and interleukin1-ß to induce hypertrophic maturation (late hypertrophic templates). Constructs were implanted in vivo and analyzed after 8 weeks. In vitro, ASC deposited cartilaginous matrix positive for glycosaminoglycans, type II collagen, and Indian hedgehog. Hypertrophic maturation induced upregulation of type X collagen, bone sialoprotein, and matrix metalloproteinase13 (MMP13). In vivo, both early and late hypertrophic templates underwent cartilage remodeling, as assessed by MMP13- and tartrate-resistant acid phosphatase-positive staining, and developed bone ossicles, including bone marrow elements, although to variable degrees of efficiency. In situ hybridization for human-specific sequences and staining with a human specific anti-CD146 antibody demonstrated the direct contribution of ASC to bone and stromal tissue formation. In conclusion, despite their debated skeletal progenitor nature, human ASC can generate bone organs through ECO when suitably primed in vitro. SIGNIFICANCE: Recapitulation of endochondral ossification (ECO) (i.e., generation of marrow-containing ossicles through a cartilage intermediate) has relevance to develop human organotypic models for bone or hematopoietic cells and to engineer grafts for bone regeneration. This study demonstrated that expanded, human adult adipose-derived stromal cells can generate ectopic bone through ECO, as previously reported for bone marrow stromal cells. This system can be used as a model in a variety of settings for mimicking ECO during development, physiology, or pathology (e.g., to investigate the role of BMPs, their receptors, and signaling pathways). The findings have also translational relevance in the field of bone regeneration, which, despite several advances in the domains of materials and surgical techniques, still faces various limitations before being introduced in the routine clinical practice.


Assuntos
Tecido Adiposo/citologia , Osso e Ossos/metabolismo , Cartilagem/metabolismo , Diferenciação Celular , Linhagem da Célula , Condrogênese , Osteogênese , Células Estromais/metabolismo , Engenharia Tecidual/métodos , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 6/metabolismo , Cartilagem/irrigação sanguínea , Células Cultivadas , Condrogênese/genética , Meios de Cultura/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Osteogênese/genética , Fenótipo , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador beta3/metabolismo
9.
EMBO Mol Med ; 7(10): 1366-84, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26323572

RESUMO

VEGF is widely investigated for therapeutic angiogenesis, but while short-term delivery is desirable for safety, it is insufficient for new vessel persistence, jeopardizing efficacy. Here, we investigated whether and how VEGF dose regulates nascent vessel stabilization, to identify novel therapeutic targets. Monoclonal populations of transduced myoblasts were used to homogeneously express specific VEGF doses in SCID mouse muscles. VEGF was abrogated after 10 and 17 days by Aflibercept treatment. Vascular stabilization was fastest with low VEGF, but delayed or prevented by higher doses, without affecting pericyte coverage. Rather, VEGF dose-dependently inhibited endothelial Semaphorin3A expression, thereby impairing recruitment of Neuropilin-1-expressing monocytes (NEM), TGF-ß1 production and endothelial SMAD2/3 activation. TGF-ß1 further initiated a feedback loop stimulating endothelial Semaphorin3A expression, thereby amplifying the stabilizing signals. Blocking experiments showed that NEM recruitment required endogenous Semaphorin3A and that TGF-ß1 was necessary to start the Semaphorin3A/NEM axis. Conversely, Semaphorin3A treatment promoted NEM recruitment and vessel stabilization despite high VEGF doses or transient adenoviral delivery. Therefore, VEGF inhibits the endothelial Semaphorin3A/NEM/TGF-ß1 paracrine axis and Semaphorin3A treatment accelerates stabilization of VEGF-induced angiogenesis.


Assuntos
Imunofilinas/metabolismo , Mioblastos , Semaforina-3A/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Indutores da Angiogênese/metabolismo , Indutores da Angiogênese/farmacologia , Animais , Camundongos , Camundongos SCID , Mioblastos/metabolismo , Mioblastos/fisiologia , Comunicação Parácrina , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
10.
Tissue Eng Part A ; 20(5-6): 1081-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24164328

RESUMO

In primary human bone marrow cultures, the initial adherent cell fraction has been shown to provide a microenvironment for self-renewal of primitive non-adherent mesenchymal progenitors (non-adherent progenitors of bone marrow stroma [BM-NAMP]), with increased differentiation potential compared to adherent colony-forming units-fibroblast (CFU-f). The present study investigates whether NAMP exist also in cultures of stromal vascular fraction (SVF) cells derived from human adipose tissue. Adipose-tissue NAMP (AT-NAMP) were shown to be stably non-adherent and their number correlated with the number of the initial adhering CFU-f. Unlike BM-NAMP, AT-NAMP did not propagate in suspension in serial replating experiments and the number of colonies steadily decreased with each replating step. However, when AT-NAMP were kept on the initially adhering SVF cells, they could significantly expand without loss of clonogenic, proliferation, and differentiation potential. Although AT-NAMP progeny differentiated into mesodermal lineages similar to that of adherent CFU-f, it was enriched in early mesenchymal progenitor populations, characterized by increased expression of SSEA-4 and CD146. Furthermore, FGF-2 supported AT-NAMP survival and could not be replaced by another mitogenic factor, such as platelet derived growth factor BB. In conclusion, these data suggest that the SVF adherent fraction provides niche signals that regulate the expansion of adipose non-adherent mesenchymal progenitors with the maintenance of their potency. The biological differences described between BM- and AT-NAMP further qualify the properties of the stroma from different tissues and will be relevant for the selection of a cell source for specific regeneration strategies.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Mesenquimais/citologia , Adipogenia , Adulto , Idoso , Adesão Celular , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Células Clonais , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Pessoa de Meia-Idade , Osteogênese , Células Estromais/citologia , Frações Subcelulares/metabolismo , Adulto Jovem
11.
Biomaterials ; 34(21): 5025-35, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23566801

RESUMO

Rapid vascularisation of tissue-engineered osteogenic grafts is a major obstacle in the development of regenerative medicine approaches for bone repair. Vascular endothelial growth factor (VEGF) is the master regulator of vascular growth. We investigated a cell-based gene therapy approach to generate osteogenic grafts with an increased vascularization potential in an ectopic nude rat model in vivo, by genetically modifying human bone marrow-derived stromal/stem cells (BMSC) to express rat VEGF. BMSC were loaded onto silicate-substituted apatite granules, which are a clinically established osteo-conductive material. Eight weeks after implantation, the vascular density of constructs seeded with VEGF-BMSC was 3-fold greater than with control cells, consisting of physiologically structured vascular networks with both conductance vessels and capillaries. However, VEGF specifically caused a global reduction in bone quantity, which consisted of thin trabeculae of immature matrix. VEGF did not impair BMSC engraftment in vivo, but strongly increased the recruitment of TRAP- and Cathepsin K-positive osteoclasts. These data suggest that VEGF over-expression is effective to improve the vascularization of osteogenic grafts, but also has the potential to disrupt bone homoeostasis towards excessive degradation, posing a challenge to its clinical application in bone tissue engineering.


Assuntos
Reabsorção Óssea/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Osteogênese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fosfatase Ácida/metabolismo , Animais , Células da Medula Óssea/metabolismo , Matriz Óssea/metabolismo , Catepsina K/metabolismo , Proliferação de Células , Sobrevivência Celular , Humanos , Isoenzimas/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoclastos/patologia , Ratos , Fosfatase Ácida Resistente a Tartarato
12.
World J Stem Cells ; 5(1): 1-8, 2013 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-23362435

RESUMO

Human adipose tissue obtained by liposuction is easily accessible and an abundant potential source of autologous cells for regenerative medicine applications. After digestion of the tissue and removal of differentiated adipocytes, the so-called stromal vascular fraction (SVF) of adipose, a mix of various cell types, is obtained. SVF contains mesenchymal fibroblastic cells, able to adhere to culture plastic and to generate large colonies in vitro, that closely resemble bone marrow-derived colony forming units-fibroblastic, and whose expanded progeny, adipose mesenchymal stem/stromal cells (ASC), show strong similarities with bone marrow mesenchymal stem cells. The sialomucin CD34, which is well known as a hematopoietic stem cell marker, is also expressed by ASC in native adipose tissue but its expression is gradually lost upon standard ASC expansion in vitro. Surprisingly little is known about the functional role of CD34 in the biology and tissue forming capacity of SVF cells and ASC. The present editorial provides a short introduction to the CD34 family of sialomucins and reviews the data from the literature concerning expression and function of these proteins in SVF cells and their in vitro expanded progeny.

13.
Stem Cells ; 30(7): 1455-64, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22495904

RESUMO

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


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Adulto , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/genética , Citometria de Fluxo , Humanos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
14.
Biomaterials ; 32(2): 321-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20952054

RESUMO

In the bone marrow, specialized microenvironments, called niches, regulate hematopoietic stem cell (HSC) maintenance and function through a complex crosstalk between different cell types. Although in vivo studies have been instrumental to elucidate some of the mechanisms by which niches exert their function, the establishment of an in vitro model that recapitulates the fundamental interactions of the niche components in a controlled setting would be of great benefit. We have previously shown that freshly harvested bone marrow- or adipose tissue-derived cells can be cultured under perfusion within porous scaffolds, allowing the formation of an organized 3D stromal tissue, composed by mesenchymal and endothelial progenitors and able to support hematopoiesis. Here we describe 3D scaffold-based perfusion systems as potential models to reconstruct ex vivo the bone marrow stem cell niche. We discuss how several culture parameters, including scaffold properties, cellular makeup and molecular signals, can be varied and controlled to investigate the role of specific cues in affecting HSC fate. We then provide a perspective of how the system could be exploited to improve stem cell-based therapies and how the model can be extended toward the engineering of other specialized stromal niches.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Nicho de Células-Tronco/citologia , Alicerces Teciduais , Animais , Humanos
15.
Arthritis Rheum ; 62(12): 3815-25, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20824797

RESUMO

OBJECTIVE: To document the specificity and the mechanism of induction of a novel class II major histocompatibility complex (MHC) antigen by mitogenic growth factors in human mesenchymal stem cells (MSCs) expanded in vitro for translational applications. METHODS: Expression of class II MHC molecules was measured in human MSCs and differentiated cells expanded in the presence of fibroblast growth factor 2 (FGF-2), platelet-derived growth factor BB (PDGF-BB), human platelet lysate, or interferon-γ (IFNγ). The roles of cell proliferation and growth factor-induced signaling pathways were investigated as well as the class II MHC assembly machinery and functional capacity. RESULTS: FGF-2 and, to a lesser extent, PDGF-BB induced in adult human MSCs the expression of HLA-DR (normally induced by inflammatory cytokines), which was able to stimulate CD4+ T cells via superantigen binding. In contrast to IFNγ, FGF induced HLA-DR expression only in human MSCs proliferating under its mitogenic effect and not in mouse MSCs or in differentiated human cells. Although it induced cell proliferation, human platelet lysate did not cause HLA-DR expression in human MSCs. HLA-DR expression occurred following FGF-specific binding to its receptor(s), mainly FGF receptor 1, without inducing IFNγ or tumor necrosis factor α expression. Both MAPK/ERK-1/2 and phosphatidylinositol 3-kinase/Akt controlled cell proliferation and HLA-DR expression, but only MAPK/ERK-1/2 controlled the induction of the class II MHC transcription activator protein CIITA, the major determinant of HLA-DR transcription. CONCLUSION: The induction of functional HLA-DR in proliferating progenitor MSCs is a property of human MSCs that have been expanded with mitogenic growth factors. This has potential biologic significance in the regulation and/or protection of progenitor cell subpopulations under sustained mitogenic proliferation and needs to be taken into account when expanding MSCs for use in in vivo applications.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Células Cultivadas , Cromonas/farmacologia , Flavonoides/farmacologia , Antígenos HLA-DR/metabolismo , Humanos , Interferon gama/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Morfolinas/farmacologia , Proteínas Nucleares/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Quinoxalinas/farmacologia , Tiazolidinedionas/farmacologia , Transativadores/metabolismo
16.
Clin Orthop Relat Res ; (435): 62-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15930922

RESUMO

Early vascular invasion is a key factor in bone allograft incorporation. It may reduce the complications related to slow and incomplete bone integration. Bone-marrow-derived stromal stem cells associated with platelet-rich plasma are potent angiogenic inducers proven to release vascular endothelial growth factor. Our goal was to test whether the combination of stromal stem cells and platelet-rich plasma is able to increase massive allograft integration in a large animal model with sacrifice at 4 months. A critical defect was made in the mid-diaphysis of the metatarsal bone of 10 sheep; the study group received an allograft plus stromal stem cells, platelet-rich plasma, and collagen (six animals) and the control group received only the allograft (four animals). Investigation was done with radiographs, mechanical tests and histomorphometric analysis, including new vascularization. Results showed substantial new bone formation in the allograft of the study group. Bone formation is correlated with better vascular invasion and remodeling of the graft in the study group. These results confirm the key role played by stromal stem cells and platelet-rich plasma in bone repair. Further studies are needed to better define the role stromal stem cells play when implanted alone.


Assuntos
Plaquetas/fisiologia , Transplante Ósseo/métodos , Osso e Ossos/irrigação sanguínea , Osseointegração/fisiologia , Transplante de Células-Tronco/métodos , Células Estromais/transplante , Cicatrização/fisiologia , Animais , Fenômenos Biomecânicos , Células Cultivadas , Método de Monte Carlo , Plasma , Distribuição Aleatória , Ovinos , Estatísticas não Paramétricas , Torque , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA