Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Stem Cells ; 33(9): 2726-37, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25969127

RESUMO

Mesenchymal stem cells (MSCs) are currently under investigation as tools to preserve cardiac structure and function following acute myocardial infarction (AMI). However, concerns have emerged regarding safety of acute intracoronary (IC) MSC delivery. This study aimed to characterize innate prothrombotic activity of MSC and identify means of its mitigation toward safe and efficacious therapeutic IC MSC delivery post-AMI. Expression of the initiator of the coagulation cascade tissue factor (TF) on MSC was detected and quantified by immunofluorescence, FACS, and immunoblotting. MSC-derived TF antigen was catalytically active and capable of supporting thrombin generation in vitro. Addition of MSCs to whole citrated blood enhanced platelet thrombus deposition on collagen at arterial shear, an effect abolished by heparin coadministration. In a porcine AMI model, IC infusion of 25 × 10(6) MSC during reperfusion was associated with a decrease in coronary flow reserve but not when coadministered with an antithrombin agent (heparin). Heparin reduced MSC-associated thrombosis incorporating platelets and VWF within the microvasculature. Heparin-assisted therapeutic MSC delivery also reduced apoptosis in the infarct border zone at 24 hours, significantly improved infarct size, left ventricular (LV) ejection fraction, LV volumes, wall motion, and attenuated histologic evidence of scar formation at 6 weeks post-AMI. Heparin alone or heparin-assisted fibroblast control cell delivery had no such effect. Procoagulant TF activity of therapeutic MSCs is associated with reductions in myocardial perfusion when delivered IC may be successfully managed by heparin coadministration. This study highlights an important mechanistic insight into safety concerns associated with therapeutic IC MSC delivery for AMI.


Assuntos
Vasos Coronários/metabolismo , Fibrinolíticos/uso terapêutico , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Microvasos/metabolismo , Tromboplastina/metabolismo , Animais , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Medula Óssea/metabolismo , Células Cultivadas , Vasos Coronários/patologia , Feminino , Fibrinolíticos/farmacologia , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Microvasos/efeitos dos fármacos , Microvasos/patologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Suínos
2.
Macromol Rapid Commun ; 36(21): 1897-1901, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26301714

RESUMO

This communication outlines the advances made in the development of thermoresponsive substrates for human mesenchymal stem cell (hMSC) expansion and subsequent controlled specific and multilineage differentiation from a previous study performed by this group. Previously, the development of an inexpensive and technically accessible method for hMSC expansion and harvesting was reported, using the solvent casting deposition method and thermoresponsive poly(N-isopropylacrylamide). Here, the logical continuation of this work is reported with the multipassage expansion of hMSCs with phenotypic maintenance followed by induced adipogenic, osteogenic, and chondrogenic differentiation. Interestingly, 1 µm thick solvent cast films are not only capable of hosting an expanding population of phenotypically preserved hMSCs similar to tissue culture plastic controls, but also the cells detached via temperature control better maintain their ability to differentiate compared to conventionally trypsinized cells. This approach to hMSC expansion and differentiation can be highly attractive to stem cell researchers where clinical therapies have seen a collective deviation away from the employment of animal derived products such as proteolytic trypsin.

3.
Biochem Biophys Res Commun ; 435(4): 574-9, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23685140

RESUMO

Mesenchymal Stem Cells are known to engraft and integrate into the architecture of colorectal tumours, with little known regarding their fate following engraftment. This study aimed to investigate mediators of Mesenchymal Stem Cell (MSC) and colon cancer cell (CCC) interactions. Mesenchymal Stem Cells and colon cancer cells (HT29 and HCT-116) were cultured individually or in co-culture on 3-dimensional scaffolds. Conditioned media containing all secreted factors was harvested at day 1, 3 and 7. Chemokine secretion and expression were analyzed by Chemi-array, ELISA (Macrophage migration inhibitory factor (MIF), plasminogen activator inhibitor type 1 (PAI-1)) and RQ-PCR. Colon cancer cell migration and proliferation in response to recombinant PAI-1, MSCs and MSCs+antibody to PAI-1 was analyzed using Transwell inserts and an MTS proliferation assay respectively. Chemi-array revealed secretion of a wide range of factors by each cell population, including PAI-1 and MIF. ELISA analysis revealed Mesenchymal Stem Cells to secrete the highest levels of PAI-1 (MSC mean 10.6 ng/mL, CCC mean 1.01 ng/mL), while colon cancer cells were the principal source of MIF. MSC-secreted PAI-1 stimulated significant migration of both CCC lines, with an antibody to the chemokine shown to block this effect (67-88% blocking,). A cell-line dependant effect on CCC proliferation was shown for Mesenchymal Stem Cell-secreted PAI-1 with HCT-116 cells showing decreased proliferation at all concentrations, and HT29 cells showing increased proliferation in the presence of higher PAI-1 levels. This is the first study to identify PAI-1 as an important mediator of Mesenchymal Stem Cell/colon cancer cell interactions and highlights the significant functional impact of Mesenchymal Stem Cell-secreted PAI-1 on colon cancer cells.


Assuntos
Comunicação Celular , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Células-Tronco Mesenquimais/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos
4.
Eur J Immunol ; 41(10): 2840-51, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21710489

RESUMO

Mesenchymal stem cells (MSCs) inhibit T-cell activation and proliferation but their effects on individual T-cell-effector pathways and on memory versus naïve T cells remain unclear. MSC influence on the differentiation of naïve and memory CD4(+) T cells toward the Th17 phenotype was examined. CD4(+) T cells exposed to Th17-skewing conditions exhibited reduced CD25 and IL-17A expression following MSC co-culture. Inhibition of IL-17A production persisted upon re-stimulation in the absence of MSCs. These effects were attenuated when cell-cell contact was prevented. Th17 cultures from highly purified naïve- and memory-phenotype responders were similarly inhibited. Th17 inhibition by MSCs was reversed by indomethacin and a selective COX-2 inhibitor. Media from MSC/Th17 co-cultures contained increased prostaglandin E2 (PGE2) levels and potently suppressed Th17 differentiation in fresh cultures. MSC-mediated Th17 inhibition was reversed by a selective EP4 antagonist and was mimicked by synthetic PGE2 and a selective EP4 agonist. Activation-induced IL-17A secretion by naturally occurring, effector-memory Th17 cells from a urinary obstruction model was also inhibited by MSC co-culture in a COX-dependent manner. Overall, MSCs potently inhibit Th17 differentiation from naïve and memory T-cell precursors and inhibit naturally-occurring Th17 cells derived from a site of inflammation. Suppression entails cell-contact-dependent COX-2 induction resulting in direct Th17 inhibition by PGE2 via EP4.


Assuntos
Dinoprostona/metabolismo , Células-Tronco Mesenquimais/fisiologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/biossíntese , Feminino , Citometria de Fluxo , Indometacina/farmacologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/biossíntese , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Ativação Linfocitária , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Receptores de Prostaglandina E Subtipo EP4/agonistas , Células Th17/efeitos dos fármacos
5.
Stem Cells ; 29(7): 1149-57, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21608083

RESUMO

Mesenchymal Stem Cells (MSCs) migrate specifically to tumors in vivo, and coupled with their capacity to bypass immune surveillance, are attractive vehicles for tumor-targeted delivery of therapeutic agents. This study aimed to introduce MSC-mediated expression of the sodium iodide symporter (NIS) for imaging and therapy of breast cancer. Tumor bearing animals received an intravenous or intratumoral injection of NIS expressing MSCs (MSC-NIS), followed by (99m) Technetium pertechnetate imaging 3-14 days later using a BazookaSPECT γ-camera. Tissue was harvested for analysis of human NIS (hNIS) expression by relative quantitative-polymerase chain reaction. Therapy animals received an i.p. injection of (131) I or saline 14 days after injection of MSC-NIS, and tumor volume was monitored for 8 weeks. After injection of MSC-NIS, BazookaSPECT imaging revealed an image of animal intestines and chest area at day 3, along with a visible weak tumor image. By day 14, the tumor was visible with a significant reduction in radionuclide accumulation in nontarget tissue observed. hNIS gene expression was detected in the intestines, heart, lungs, and tumors at early time points but later depleted in nontarget tissues and persisted at the tumor site. Based on imaging/biodistribution data, animals received a therapeutic dose of (131) I 14 days after MSC-NIS injection. This resulted in a significant reduction in tumor growth (mean ± SEM, 236 ± 62 mm(3) vs. 665 ± 204 mm(3) in controls). The ability to track MSC migration and transgene expression noninvasively in real time before therapy is a major advantage to this strategy. This promising data supports the feasibility of this approach as a novel therapy for breast cancer.


Assuntos
Neoplasias da Mama/terapia , Terapia Genética/métodos , Células-Tronco Mesenquimais/fisiologia , Simportadores/biossíntese , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Radioisótopos do Iodo/farmacocinética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase , Cintilografia , Simportadores/genética , Distribuição Tecidual , Transfecção
6.
Cytotherapy ; 12(3): 313-25, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20370348

RESUMO

BACKGROUND AIMS: Spinal cord injury is a devastating injury that impacts drastically on the victim's quality of life. Stem cells have been proposed as a therapeutic strategy. Neural stem (NS) cells have been harvested from embryonic mouse forebrain and cultured as adherent cells. These NS cells express markers of neurogenic radial glia. METHODS: Mouse NS cells expressing green fluorescent protein (GFP) were transplanted into immunosupressed rat spinal cords following moderate contusion injury at T9. Animals were left for 2 and 6 weeks then spinal cords were fixed, cryosectioned and analyzed. Stereologic methods were used to estimate the volume and cellular environment of the lesions. Engraftment, migration and differentiation of NS cells were also examined. RESULTS: NS cells integrated well into host tissue and appeared to migrate toward the lesion site. They expressed markers of neurons, astrocytes and oligodendrocytes at 2 weeks post-transplantation and markers of neurons and astrocytes at the 6-week time-point. NS cells appeared to have a similar morphologic phenotype to radial glia, in particular at the pial surface. CONCLUSIONS: Although no functional recovery was observed using the Basso Beattie Bresnahan (BBB) locomotor rating scale, NS cells are a potential cellular therapy for treatment of injured spinal cord. They may be used as delivery vehicles for therapeutic proteins because they show an ability to migrate toward the site of a lesion. They may also be used to replace lost or damaged neurons and oligodendrocytes.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Neurônios , Traumatismos da Medula Espinal , Medula Espinal , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Comportamento Animal/fisiologia , Biomarcadores/metabolismo , Sobrevivência Celular , Células Cultivadas , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Neurônios/transplante , Distribuição Aleatória , Ratos , Medula Espinal/citologia , Medula Espinal/patologia , Medula Espinal/transplante , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/cirurgia , Células-Tronco/citologia
7.
Int J Cancer ; 124(2): 326-32, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19003962

RESUMO

Over 70% of patients with advanced breast cancer will develop bone metastases for which there is no cure. Mesenchymal Stem Cells (MSCs) and their derivative osteoblasts are subpopulations of cells within the bone marrow environment, postulated as potential interacting targets for disseminating cancer cells because of their ability to secrete a range of chemokines. This study aimed to investigate chemokine secretion throughout MSC differentiation into osteoblasts and their effect on the breast cancer cells. Primary MSCs and osteoblast progenitors were cultured in appropriate conditions to induce differentiation into mature osteoblasts. Chemokines secreted throughout differentiation were detected using ChemiArray and ELISA. Migration of breast cancer cells in response to the bone-derived cells was quantified using Transwell inserts. Breast cancer cells were cocultured with MSCs, retrieved using magnetic beads, and changes in CCL2 expression were analyzed. MSCs secreted a range of factors including IL-6, TIMP-1 and CCL2, the range and level of which changed throughout differentiation. CCL2 secretion by MSCs increased significantly above control cells as they differentiated into mature osteoblasts (p<0.05). The bone-derived cells stimulated migration of breast cancer cells, and this was inhibited (21-50%) in the presence of a CCL2 antibody. CCL2 gene expression in breast cancer cells was upregulated following direct coculture with MSCs. The varying levels of chemokines secreted throughout MSC differentiation may play an important role in supporting tumor cell homing and progression. These results further highlight the distinct effect MSCs have on breast cancer cells and their potential importance in supporting development of metastases.


Assuntos
Neoplasias da Mama/metabolismo , Quimiocinas/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoblastos/metabolismo , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CCL2/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Modelos Biológicos , Metástase Neoplásica
8.
J Anat ; 215(3): 267-79, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19558472

RESUMO

Considerable evidence has shown that the immunosuppressant drug cyclosporin A (CsA) may have neuroprotective properties which can be exploited in the treatment of spinal cord injury. The aim of this study was to investigate the cellular environment within the spinal cord following injury and determine whether CsA has an effect on altering cellular interactions to promote a growth-permissive environment. CsA was administered to a group of rats 4 days after they endured a moderate contusion injury. Functional recovery was assessed using the Basso Beattie Bresnahan (BBB) locomotor rating scale at 3, 5 and 7 weeks post-injury. The rats were sacrificed 3 and 7 weeks post-injury and the spinal cords were sectioned, stained using histological and immunohistochemical methods and analysed. Using stereology, the lesion size and cellular environment in the CsA-treated and control groups was examined. Little difference in lesion volume was observed between the two groups. An improvement in functional recovery was observed within CsA-treated animals at 3 weeks. Although we did not see significant reduction in tissue damage, there were some notable differences in the proportion of individual cells contributing to the lesion. CsA administration may be used as a technique to control the cell population of the lesion, making it more permissive to neuronal regeneration once the ideal environment for regeneration and the effects of CsA administration at different time points post-injury have been identified.


Assuntos
Ciclosporina/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Colágeno/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Membro Posterior/fisiopatologia , Microscopia Eletrônica de Varredura/métodos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/ultraestrutura , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia
9.
JOR Spine ; 2(4): e1072, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31891121

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) are becoming an increasingly attractive option for regenerative therapies due to their availability, self-renewal capacity, multilineage potential, and anti-inflammatory properties. Clinical trials are underway to test the efficacy of stem cell-based therapies for the repair and regeneration of the degenerate intervertebral disc (IVD), a major cause of back pain. Recently, both bone marrow-derived MSCs and adipose-derived stem cells (ASCs) have been assessed for IVD therapy but there is a lack of knowledge surrounding the optimal cell source and the response of transplanted cells to the low oxygen, pro-inflammatory niche of the degenerate disc. Here, we investigated several neurovascular factors from donor-matched MSCs and ASCs that may potentiate the survival and persistence of sensory nerve fibers and blood vessels present within painful degenerate discs and their regulation by oxygen tensions and inflammatory cytokines. METHODS: Donor-matched ASCs and MSCs were conditioned with either IL-1ß or TNFα under normoxic (21% O2) or hypoxic (5% O2) conditions. Expression and secretion of several potent neurovascular factors were assessed using qRT-PCR and human magnetic Luminex assay. RESULTS: ASCs and MSCs expressed constitutive levels of key neurotrophic factors; and stimulation of ASCs with hypoxia triggered increased secretion of both angiogenic factors (Ang-2 and VEGF-A) and neurotrophic (NGF and NT-3) compared to MSCs. We also report increased transcriptional regulation of pain-associated neuropeptides in hypoxia stimulated ASCs compared to those in normoxic conditions. We demonstrate transcriptional and translational upregulation of NGF, NT-3, Ang-1, and FGF-2 in response to cytokines in ASCs in 21% and 5% O2. CONCLUSIONS: This work highlights fundamental differences between the neurovascular secretome of donor-matched ASCs and MSCs, demonstrating the importance of cell-selection for tissue specific regeneration to reduce ectopic sensory nerve and blood vessel survival and improve patient outcomes.

10.
Sci Rep ; 9(1): 13274, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31527619

RESUMO

Articular cartilage lacks an intrinsic repair capacity and due to the ability of mesenchymal stem cells (MSCs) to differentiate into chondrocytes, MSCs have been touted as a cellular source to regenerate damaged cartilage. However, a number of prevailing concerns for such a treatment remain. Generally, administration of MSCs into a cartilage defect results in poor regeneration of the damaged cartilage with the repaired cartilage consisting primarily of fibro-cartilage rather than hyaline cartilage. Methods that improve the chondrogenic potential of transplanted MSCs in vivo may be advantageous. In addition, the proclivity of MSC-derived cartilage to undergo hypertrophic differentiation or form bone in vivo also remains a clinical concern. If MSC-derived cartilage was to undergo hypertrophic differentiation in vivo, this would be deleterious in a clinical setting. This study focuses on establishing a mechanism of action by which hypoxia or low oxygen tension can be used to both enhance chondrogenesis and attenuate hypertrophic differentiation of both MSC and ATDC5 derived chondrocytes. Having elucidated a novel mechanism of action, the subsequent goals of this study were to develop an in vitro culture regime to mimic the beneficial effects of physiological low oxygen tension in a normoxic environment.


Assuntos
Anaerobiose/fisiologia , Cartilagem Articular/citologia , Hipóxia Celular/fisiologia , Condrogênese/fisiologia , Hipertrofia/prevenção & controle , Células-Tronco Mesenquimais/citologia , Animais , Linhagem Celular Tumoral , Condrócitos/citologia , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Isoquinolinas/farmacologia , Fatores de Transcrição MEF2/metabolismo , Transplante de Células-Tronco Mesenquimais , Camundongos , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia
11.
Immunol Lett ; 115(1): 50-8, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18022251

RESUMO

Mesenchymal stem cells (MSC) possess a wide range of immunosuppressive functions. Among these is the ability to inhibit CD4+ T cell proliferation. Dendritic cells (DC) play a role in initiating cell-mediated immunity; however, the immunosuppressive influence of MSC on professional antigen presenting cells remains unclear. DC exposed to TNF-alpha and cultured with murine MSC failed to show regular upregulation of maturation markers. Similarly, the presence of MSC abrogated the capacity of ovalbumin-pulsed DC to support antigen specific CD4+ T cell proliferation, or for DC to display an MHC class II- peptide complex recognizable by specific antibody. Interestingly, culture of MSC with DC resulted in reduced expression of CCR7 by DC following stimulation. Likewise, DC matured in the presence of MSC, showed significantly less migration to CCL19. In contrast, murine MSC prevented loss of expression of the tissue anchoring protein E-cadherin by DC. Modulation of DC maturation and function was not permanent and could be restored after removal of MSC. These data demonstrate that MSC modulate the three cardinal features of DC maturation, providing the first demonstration of MSC interference with DC migration.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Quimiotaxia , Células Dendríticas/imunologia , Células-Tronco Mesenquimais/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Quimiocina CCL19/imunologia , Quimiocina CCL19/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Imunidade Celular , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismo
12.
Biomaterials ; 29(11): 1610-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18194813

RESUMO

Understanding the response of mesenchymal stem cells (MSCs) to forces in the vasculature is very important in the field of cardiovascular intervention for a number of reasons. These include the development of MSC seeded tissue engineered vascular grafts, targeted or systemic delivery of MSCs in the dynamic environment of the coronary artery and understanding the potential pathological calcifying role of mechanically conditioned multipotent cells already present in the vessel wall. In vivo, cells present in the coronary artery are exposed to the primary biomechanical forces of shear stress, radial stress and hoop stress. To date, many studies have examined the effect of these stresses in isolation, thereby not presenting the complete picture. Therefore, the main aim of this study is to examine the combined role of these stresses on MSC behaviour. To this end, a bioreactor was configured to expose MSCs seeded on flexible silicone substrates to physiological forces - namely, a pulsatile pressure between 40 and 120mmHg (5.33-1.6x10(4)Pa), radial distention of 5% and a shear stress of 10dyn/cm(2) (1Pa) at frequency of 1Hz for up to 24h. Thereafter, the 'pseudovessel' was assessed for changes in morphology, orientation and expression of endothelial and smooth muscle cell (SMC) specific markers. Hematoxylin and eosin (H&E) staining revealed that MSCs exhibit a similar mechanosensitive response to that of endothelial cells (ECs); they reorientate parallel with direction of flow and have adapted their morphology to be similar to that of ECs. However, gene expression results show the cells exhibit greater levels of SMC-associated markers alpha-smooth muscle actin and calponin (p<0.05).


Assuntos
Endotélio/metabolismo , Células-Tronco Mesenquimais/citologia , Maleabilidade , Silicones , Adolescente , Adulto , Fenômenos Biomecânicos , Reatores Biológicos , Forma Celular , Células Cultivadas , Endotélio/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , RNA Mensageiro/genética
13.
Immunol Lett ; 110(2): 91-100, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17507101

RESUMO

Murine mesenchymal stem cells (MSC) have the ability to inhibit allogeneic immune responses. Two different mechanisms, either cell contact-dependent or independent, have been proposed to account for this immunosuppression. The focus of this study was to elucidate the involvement of soluble suppressive factors secreted by murine MSC in an inflammatory setting, and their role in MSC immunomodulation. In a non-inflammatory environment, bone marrow derived murine MSC constitutively expressed low levels of COX-2, PGE-2, TGF-beta1 and HGF, but not IL-10, PD-1, PD-L1 or PD-L2. These MSC were able to significantly reduce alloantigen driven proliferation in mixed lymphocyte reactions as well as mitogen driven proliferation. The pro-inflammatory cytokines IFN-gamma and TNF-alpha did not ablate MSC mediated immunosuppression. MSC expression of PGE-2, IDO and PD-L1 was differentially regulated by these cytokines. COX-2 and PGE-2 expression by MSC were upregulated by both IFN-gamma and TNF-alpha, and using a biochemical inhibitor this was shown to have an essential, non-redundant role in modulating alloantigen-driven proliferation. However, the surface expression of PD-L1 was induced by IFN-gamma but not TNF-alpha and similarly functional IDO expression was only induced by IFN-gamma stimulation. Blocking studies using neutralising antibodies and biochemical antagonists revealed that while PD-L1 induction was not essential, IDO expression was a prerequisite for IFN-gamma mediated MSC immunomodulation. These data demonstrate that murine MSC expression of immunomodulatory factors dramatically changes in a pro-inflammatory environment and that IFN-gamma in particular has an important role in regulating MSC immunomodulatory factor expression.


Assuntos
Citocinas/imunologia , Interferon gama/imunologia , Células-Tronco Mesenquimais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Células da Medula Óssea , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Feminino , Fatores Imunológicos/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indometacina/farmacologia , Interferon gama/metabolismo , Teste de Cultura Mista de Linfócitos , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Peptídeos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
J Mater Chem B ; 4(20): 3482-3489, 2016 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32263381

RESUMO

Disease-specific pluripotent stem cells can be derived through genetic manipulation of embryonic stem cells or by reprogramming somatic cells (induced pluripotent stem cells). These cells are a valuable tool to study human diseases in vitro in order to dissect their pathomechanisms and develop novel therapeutics. Although pluripotent stem cell-derived models have successfully recapitulated the abnormalities of some skeletal diseases in vitro, this field is still at its early stages, and it could greatly benefit from the direct application of biomaterial research. Biomaterial-based systems may be utilized to enhance the differentiation processes of pluripotent stem cells in order to create more homogeneous and physiologically relevant in vitro disease models. Moreover, inducing the disease phenotype may be facilitated by the guidance of biomaterials. This review presents a comprehensive summary of existing biomaterial applications in human disease modeling and their potential on skeletal disease models. By utilizing disease-specific pluripotent stem cells, current biomaterial-based systems for in vitro models could be extrapolated to study skeletal diseases in a petri dish.

15.
Stem Cells Dev ; 14(3): 252-65, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15969620

RESUMO

Herein we review recent data that support host tolerance of allogeneic adult mesenchymal stem cells (MSC). Evidence is emerging that donor MSC deploy a very powerful array of mechanisms that allow escape from host allogeneic responses. These mechanisms include limited expression of alloantigen by the stem cell and cell contact-dependent and -independent mechanisms. MSC modulate host dendritic cell and T cell function, promoting induction of suppressor or regulatory T cells. These effects are complemented by the induction of divisional arrest anergy in T cells and by stem cell production of soluble immunomodulatory factors, including interleukin-10, transforming growth factor-beta, prostaglandin E2, and hepatocyte growth factor. In addition, MSC express the enzyme indoleamine 2,3-dioxygenase, which creates a tryptophan-depleted milieu that promotes immunosuppression. We propose that these observations show striking similarity to emerging data on the maternal acceptance of the fetal allograft. This comparison suggests new approaches to determine the contribution of different mechanisms to the successful use of MSC in regenerative medicine.


Assuntos
Transplante de Tecido Fetal/imunologia , Mesoderma/imunologia , Células-Tronco/microbiologia , Adulto , Feminino , Rejeição de Enxerto/imunologia , Humanos , Complexo Principal de Histocompatibilidade , Troca Materno-Fetal/imunologia , Gravidez , Transplante Homólogo/imunologia
16.
Stem Cell Res Ther ; 6: 136, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26204937

RESUMO

INTRODUCTION: Bone marrow-derived stromal cells (BMSCs), also known as mesenchymal stem cells, are the focus of intensive efforts worldwide to elucidate their function and biology. Despite the importance of BMSC migration for their potential therapeutic uses, the mechanisms and signalling governing stem cell migration are still not fully elucidated. METHODS: We investigated and detailed the effects of MCP-1 activation on BMSCs by using inhibitors of G protein-coupled receptor alpha beta (GPCR αß), ROCK (Rho-associated, coiled-coil containing protein kinase), and PI3 kinase (PI3K). The effects of MCP-1 stimulation on intracellular signalling cascades were characterised by using immunoblotting and immunofluorescence. The effectors of MCP-1-mediated migration were investigated by using migration assays (both two-dimensional and three-dimensional) in combination with inhibitors. RESULTS: We established the kinetics of the MCP-1-activated signalling cascade and show that this cascade correlates with cell surface re-localisation of chemokine (C motif) receptor 2 (CCR2) (the MCP-1 receptor) to the cell periphery following MCP-1 stimulation. We show that MCP-1-initiated signalling is dependent on the activation of ßγ subunits from the GPCR αßγ complex. In addition, we characterise a novel role for PI3Kγ signalling for the activation of both PAK and ERK following MCP-1 stimulation. We present evidence that the Gßγ complex is responsible for PI3K/Akt, PAK, and ERK signalling induced by MCP-1 in BMSCs. Importantly, we found that, in BMSCs, inhibition of ROCK significantly inhibits MCP-1-induced chemotactic migration, in contrast to previous reports in other systems. CONCLUSIONS: Our results indicate differential chemotactic signalling in mouse BMSCs, which has important implications for the translation of in vivo mouse model findings into human trials. We identified novel components and interactions activated by MCP-1-mediated signalling, which are important for stem cell migration. This work has identified additional potential therapeutic targets that could be manipulated to improve BMSC delivery and homing.


Assuntos
Quimiotaxia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Células-Tronco Mesenquimais/fisiologia , Quinases Associadas a rho/metabolismo , Animais , Antígenos/metabolismo , Células Cultivadas , Quimiocina CCL2/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
17.
Int J Biochem Cell Biol ; 36(4): 568-84, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15010324

RESUMO

Mesenchymal stem cells (MSCs) have been isolated from bone marrow, periosteum, trabecular bone, adipose tissue, synovium, skeletal muscle and deciduous teeth. These cells have the capacity to differentiate into cells of connective tissue lineages, including bone, fat, cartilage and muscle. A great deal has been learned in recent years about the isolation and characterization of MSCs, and control of their differentiation. These cells have generated a great deal of interest because of their potential use in regenerative medicine and tissue engineering and there are some dramatic examples, derived from both pre-clinical and clinical studies, that illustrate their therapeutic value. This review summarizes recent findings regarding the potential clinical use of MSCs in cardiovascular, neural and orthopaedic applications. As new methods are developed, there are several aspects to the implanted cell-host interaction that need to be addressed before we can fully understand the underlying mechanisms. These include the host immune response to implanted cells, the homing mechanisms that guide delivered cells to a site of injury and the differentiation in vivo of implanted cells under the influence of local signals.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Antígenos de Superfície/isolamento & purificação , Medula Óssea/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular , Condrogênese , Cabras , Humanos , Células-Tronco Mesenquimais/fisiologia , Modelos Animais
18.
Novartis Found Symp ; 249: 86-96; discussion 96-102, 170-4, 239-41, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12708651

RESUMO

Mesenchymal stem cells have the capacity to differentiate into a variety of connective tissue cells including bone, cartilage, tendon, muscle and adipose tissue. These multipotent cells have been isolated from bone marrow and from other adult tissues including skeletal muscle, fat and synovium. Because of their multipotentiality and capacity for self renewal adult stem cells may represent units of active regeneration of tissues damaged as a result of trauma or disease. In certain degenerative diseases such as osteoarthritis (OA) stem cells are depleted, and have reduced proliferative capacity and reduced ability to differentiate. The delivery of stem cells to these individuals may therefore enhance repair or inhibit the progressive destruction of the joint. We have developed methods for the delivery of mesenchymal stem cell preparations taken from bone marrow to the injured knee joint. This treatment has the potential to stimulate regeneration of cartilage and retard the progressive destruction of the joint that typically occurs following injury.


Assuntos
Artropatias/cirurgia , Mesoderma/citologia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco , Adulto , Animais , Antígenos de Diferenciação/análise , Cartilagem Articular/lesões , Cartilagem Articular/patologia , Diferenciação Celular , Citometria de Fluxo , Genes Reporter , Cabras , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Meniscos Tibiais/cirurgia , Osteoartrite/patologia , Osteoartrite/cirurgia , Células Estromais/citologia
19.
J Control Release ; 179: 42-51, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24491910

RESUMO

Human mesenchymal stem cells (hMSCs) have been identified as a viable cell source for cartilage tissue engineering. However, to undergo chondrogenic differentiation hMSCs require growth factors, in particular members of the transforming growth factor beta (TGF-ß) family. While in vitro differentiation is feasible through continuous supplementation of TGF-ß3, mechanisms to control and drive hMSCs down the chondrogenic lineage in their native microenvironment remain a significant challenge. The release of TGF-ß3 from an injectable microsphere composed of the cartilage-associated extracellular matrix molecule hyaluronan represents a readily translatable approach for in situ differentiation of hMSCs for cartilage repair. In this study, chondromimetic hyaluronan microspheres were used as a growth factor delivery source for hMSC chondrogenesis. Cellular compatibility of the microspheres (1.2 and 14.1 µm) with hMSCs was shown and release of TGF-ß3 from the most promising 14.1 µm microspheres to control differentiation of hMSCs was evaluated. Enhanced accumulation of cartilage-associated glycosaminoglycans by hMSCs incubated with TGF-ß3-loaded microspheres was seen and positive staining for collagen type II and proteoglycan confirmed successful in vitro chondrogenesis. Gene expression analysis showed significantly increased expression of the chondrocyte-associated genes, collagen type II and aggrecan. This delivery platform resulted in significantly less collagen type X expression, suggesting the generation of a more stable cartilage phenotype. When evaluated in an ex vivo osteoarthritic cartilage model, implanted hMSCs with TGF-ß3-loaded HA microspheres were detected within cartilage fibrillations and increased proteoglycan staining was seen in the tissue. In summary, data presented here demonstrate that TGF-ß3-bound hyaluronan microspheres provide a suitable delivery system for induction of hMSC chondrogenesis and their use may represent a clinically feasible tissue engineering approach for the treatment of articular cartilage defects.


Assuntos
Biomimética , Diferenciação Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Portadores de Fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Engenharia Tecidual , Fator de Crescimento Transformador beta3/farmacologia , Adolescente , Adulto , Agrecanas/genética , Agrecanas/metabolismo , Animais , Linhagem Celular , Condrócitos/metabolismo , Condrócitos/transplante , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Humanos , Ácido Hialurônico/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Microesferas , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/terapia , Fenótipo , Fatores de Tempo , Fator de Crescimento Transformador beta3/toxicidade , Adulto Jovem
20.
Stem Cell Rev Rep ; 9(2): 148-57, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23354660

RESUMO

The facile regeneration of undifferentiated human mesenchymal stem cells (hMSCs) from thermoresponsive surfaces facilitates the collection of stem cells avoiding the use of animal derived cell detachment agents commonly used in cell culture. This communication proposes a procedure to fabricate coatings from commercially available pNIPAm which is both affordable and a significant simplification on alternative approaches used elsewhere. Solvent casting was used to produce films in the micrometer range and successful cell adhesion and proliferation was highly dependent on the thickness of the coating produced with 1 µm thick coatings supporting cells to confluence. 3T3 cell sheets and hMSCs were successfully detached from the cast coatings upon temperature reduction. Furthermore, results indicate that the hMSCs remained undifferentiated as the surface receptor profile of hMSCs was not altered when cells were detached in this manner.


Assuntos
Resinas Acrílicas/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Resinas Acrílicas/química , Animais , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Células NIH 3T3 , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA