Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Stem Cell Res Ther ; 15(1): 178, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38886845

RESUMEN

BACKGROUND: Bone marrow stromal cells (BMSCs) are highly heterogeneous, which may reflect their diverse biological functions, including tissue maintenance, haematopoietic support and immune control. The current understanding of the mechanisms that drive the onset and resolution of heterogeneity, and how BMSCs influence other cells in their environment is limited. Here, we determined how the secretome and importantly the extracellular matrix of BMSCs can influence cellular phenotype. METHODS: We used two immortalised clonal BMSC lines isolated from the same heterogeneous culture as model stromal subtypes with distinct phenotypic traits; a multipotent stem-cell-like stromal line (Y201) and a nullipotent non-stem cell stromal line (Y202), isolated from the same donor BMSC pool. Label-free quantitative phase imaging was used to track cell morphology and migration of the BMSC lines over 96 h in colony-forming assays. We quantified the secreted factors of each cell line by mass spectrometry and confirmed presence of proteins in human bone marrow by immunofluorescence. RESULTS: Transfer of secreted signals from a stem cell to a non-stem cell resulted in a change in morphology and enhanced migration to more closely match stem cell-like features. Mass spectrometry analysis revealed a significant enrichment of extracellular matrix (ECM) proteins in the Y201 stem cell secretome compared to Y202 stromal cells. We confirmed that Y201 produced a more robust ECM in culture compared to Y202. Growth of Y202 on ECM produced by Y201 or Y202 restored migration and fibroblastic morphology, suggesting that it is the deficiency of ECM production that contributes to its phenotype. The proteins periostin and aggrecan, were detected at 71- and 104-fold higher levels in the Y201 versus Y202 secretome and were subsequently identified by immunofluorescence at rare sites on the endosteal surfaces of mouse and human bone, underlying CD271-positive stromal cells. These proteins may represent key non-cellular components of the microenvironment for bona-fide stem cells important for cell maintenance and phenotype in vivo. CONCLUSIONS: We identified plasticity in BMSC morphology and migratory characteristics that can be modified through secreted proteins, particularly from multipotent stem cells. Overall, we demonstrate the importance of specific ECM proteins in co-ordination of cellular phenotype and highlight how non-cellular components of the BMSC microenvironment may provide insights into cell population heterogeneity and the role of BMSCs in health and disease.


Asunto(s)
Matriz Extracelular , Células Madre Mesenquimatosas , Fenotipo , Humanos , Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Células del Estroma/metabolismo , Células del Estroma/citología , Línea Celular
2.
Stem Cells Transl Med ; 12(11): 745-757, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37713249

RESUMEN

Pharmacopoeial standards ensure quality control of established medicines. It is widely believed that translation of cell therapy medicines will be facilitated by defining and adopting relevant standards. Mesenchymal stromal cells (MSCs) are used extensively for multiple indications in regenerative medicine. They are highly heterogeneous in terms of their biological characteristics and their mechanisms of action, making standardization a challenging undertaking. Furthermore, the use of MSCs in therapy appears to attract diverse views, ranging from concern and caution to enthusiastic positivity. We conducted semi-structured interviews with 20 expert stakeholders from academia, industry, regulatory agencies, non-governmental organizations and clinicians to explore their views, experiences, recommendations, and concerns regarding standardization of MSCs. Qualitative thematic analysis of transcribed records led to development of a consensus framework, which identified 5 key themes to facilitate exploration of the interviews' content. On the basis of our findings, we conclude that (1) there is undoubtedly an appetite for standardization, particularly in development of assays that enable comparison or benchmarking across manufacturers, processes, and cell sources; (2) stakeholder groups are not homogeneous in their concerns and attitudes; (3) careful consideration must be given to the points along the development timeline at which different standardization approaches could be beneficial; and (4) the roles of standards could be promoted further for specific aspects of advanced therapy medicinal product (ATMP) development and regulation such as qualification of decentralized manufacturing sites. A unified cross-stakeholder approach will help to advance MSC therapeutics and other cell therapy medicines.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Estándares de Referencia , Control de Calidad , Actitud
3.
Front Immunol ; 13: 903796, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35734183

RESUMEN

Heterogeneity of bone marrow mesenchymal stromal cells (MSCs, frequently referred to as "mesenchymal stem cells") clouds biological understanding and hampers their clinical development. In MSC cultures most commonly used in research and therapy, we have identified an MSC subtype characterized by CD317 expression (CD317pos (29.77 ± 3.00% of the total MSC population), comprising CD317dim (28.10 ± 4.60%) and CD317bright (1.67 ± 0.58%) MSCs) and a constitutive interferon signature linked to human disease. We demonstrate that CD317pos MSCs induced cutaneous tissue damage when applied a skin explant model of inflammation, whereas CD317neg MSCs had no effect. Only CD317neg MSCs were able to suppress proliferative cycles of activated human T cells in vitro, whilst CD317pos MSCs increased polarization towards pro-inflammatory Th1 cells and CD317neg cell lines did not. Using an in vivo peritonitis model, we found that CD317neg and CD317pos MSCs suppressed leukocyte recruitment but only CD317neg MSCs suppressed macrophage numbers. Using MSC-loaded scaffolds implanted subcutaneously in immunocompromised mice we were able to observe tissue generation and blood vessel formation with CD317neg MSC lines, but not CD317pos MSC lines. Our evidence is consistent with the identification of an immune stromal cell, which is likely to contribute to specific physiological and pathological functions and influence clinical outcome of therapeutic MSCs.


Asunto(s)
Células Madre Mesenquimatosas , Animales , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Transducción de Señal , Células del Estroma , Células TH1
4.
Blood Adv ; 5(23): 4877-4889, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34428275

RESUMEN

Immune thrombocytopenia (ITP) is an acquired autoimmune condition characterized by both reduced platelet production and the destruction of functionally normal platelets by sustained attack from the immune system. However, the effect of prolonged ITP on the more immature hematopoietic progenitors remains an open area of investigation. By using a murine in vivo model of extended ITP, we revealed that ITP progression drives considerable progenitor expansion and bone marrow (BM) remodeling. Single-cell assays using Lin-Sca1+c-Kit+CD48-CD150+ long-term hematopoietic stem cells (LT-HSCs) revealed elevated LT-HSC activation and proliferation in vitro. However, the increased activation did not come at the expense of LT-HSC functionality as measured by in vivo serial transplantations. ITP progression was associated with considerable BM vasodilation and angiogenesis, as well as a twofold increase in the local production of CXCL12, a cytokine essential for LT-HSC function and BM homing expressed at high levels by LepR+ BM stromal cells. This was associated with a 1.5-fold increase in LepR+ BM stromal cells and a 5.5-fold improvement in progenitor homing to the BM. The increase in stromal cells was transient and reverted back to baseline after platelet count returned to normal, but the vasculature changes in the BM persisted. Together, our data demonstrate that LT-HSCs expand in response to ITP and that LT-HSC functionality during sustained hematopoietic stress is maintained through an adapting BM microenvironment.


Asunto(s)
Médula Ósea , Púrpura Trombocitopénica Idiopática , Animales , Hematopoyesis , Células Madre Hematopoyéticas , Ratones , Ratones Endogámicos C57BL
5.
Stem Cell Res Ther ; 12(1): 360, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158116

RESUMEN

BACKGROUND: Mesenchymal stem or stromal cells are the most widely used cell therapy to date. They are heterogeneous, with variations in growth potential, differentiation capacity and protein expression profile depending on tissue source and production process. Nomenclature and defining characteristics have been debated for almost 20 years, yet the generic term 'MSC' is used to cover a wide range of cellular phenotypes. Against a documented lack of definition of cellular populations used in clinical trials, our study evaluated the extent of characterisation of the cellular population or study drug. METHODS: A literature search of clinical trials involving mesenchymal stem/stromal cells was refined to 84 papers upon application of pre-defined inclusion/exclusion criteria. Data were extracted covering background trial information including location, phase, indication, tissue source and details of clinical cell population characterisation (expression of surface markers, viability, differentiation assays and potency/functionality assays). Descriptive statistics were applied, and tests of association between groups were explored using Fisher's exact test for count data with simulated p value. RESULTS: Twenty-eight studies (33.3%) include no characterisation data. Forty-five (53.6%) reported average values per marker for all cell lots used in the trial, and 11 (13.1%) studies included individual values per cell lot. Viability was reported in 57% of studies. Differentiation was discussed: osteogenesis (29% of papers), adipogenesis (27%), and chondrogenesis (20%) and other functional assays arose in 7 papers (8%). The extent of characterisation was not related to the clinical phase of development. Assessment of functionality was very limited and did not always relate to the likely mechanism of action. CONCLUSIONS: The extent of characterisation was poor and variable. Our findings concur with those in other fields including bone marrow aspirate and platelet-rich plasma therapy. We discuss the potential implications of these findings for the use of mesenchymal stem or stromal cells in regenerative medicine, and the importance of characterisation for transparency and comparability of literature.


Asunto(s)
Células Madre Mesenquimatosas , Adipogénesis , Células de la Médula Ósea , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrogénesis , Osteogénesis
6.
Mater Sci Eng C Mater Biol Appl ; 89: 149-159, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29752083

RESUMEN

This paper presents an investigation of how different culture media (i.e. basal and osteogenic media) affect the nanomechanical properties and microstructure of the mineralized matrix produced by the human mesenchymal stem cell line Y201, from both an experimental and theoretical approach. A bone nodule (i.e. mineralized matrix) cultured from basal medium shows a more anisotropic microstructure compared to its counterpart cultured from an osteogenic medium. As confirmed by finite element simulations, this anisotropic microstructure explains the bimodal distribution of the corresponding mechanical properties very well. The overall nanomechanical response of the bone nodule from the osteogenic medium is poorer compared to its counterpart from the basal medium. The bone nodules, from both basal and osteogenic media, have shown reverse aging effects in terms of mechanical properties. These are possibly due to the fact that cell proliferation outcompetes the mineralization process.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Matriz Extracelular/metabolismo , Modelos Biológicos , Nanoestructuras/química , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Línea Celular , Módulo de Elasticidad , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Análisis de Elementos Finitos , Humanos , Células Madre Mesenquimatosas/citología , Osteogénesis , Propiedades de Superficie
7.
Int J Numer Method Biomed Eng ; 32(3): e02735, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26198466

RESUMEN

Multiple myeloma bone disease is devastating for patients and a major cause of morbidity. The disease leads to bone destruction by inhibiting osteoblast activity while stimulating osteoclast activity. Recent advances in multiple myeloma research have improved our understanding of the pathogenesis of multiple myeloma-induced bone disease and suggest several potential therapeutic strategies. However, the effectiveness of some potential therapeutic strategies still requires further investigation and optimization. In this paper, a recently developed mathematical model is extended to mimic and then evaluate three therapies of the disease, namely: bisphosphonates, bortezomib and TGF-ß inhibition. The model suggests that bisphosphonates and bortezomib treatments not only inhibit bone destruction, but also reduce the viability of myeloma cells. This contributes to the current debate as to whether bisphosphonate therapy has an anti-tumour effect. On the other hand, the analyses indicate that treatments designed to inhibit TGF-ß do not reduce bone destruction, although it appears that they might reduce the viability of myeloma cells, which again contributes to the current controversy regarding the efficacy of TGF-ß inhibition in multiple myeloma-induced bone disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/etiología , Mieloma Múltiple/complicaciones , Mieloma Múltiple/tratamiento farmacológico , Interfaz Usuario-Computador , Antineoplásicos/farmacología , Bortezomib/farmacología , Bortezomib/uso terapéutico , Simulación por Computador , Humanos , Modelos Biológicos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
8.
Biomacromolecules ; 17(3): 723-34, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26686360

RESUMEN

There is a lack of hydrogel materials whose properties can be tuned at the point of use. Biological hydrogels, such as collagen, gelate at physiological temperatures; however, they are not always ideal as scaffolds because of their low mechanical strength. Their mechanics can be improved through cross-linking and chemical modification, but these methods still require further synthesis. We have demonstrated that by combining collagen with a thermoresponsive polymer, poly(N-isopropylacrylamide) (PNIPAM), the mechanical properties can be improved while maintaining cytocompatibility. Furthermore, different concentrations of this polymer led to a range of hydrogels with shear moduli ranging from 10(5) Pa down to less than 10(2) Pa, similar to the soft tissues in the body. In addition to variable mechanical properties, the hydrogel blends have a range of micron-scale structures and porosities, which caused adipose-derived stromal cells (ADSCs) to adopt different morphologies when encapsulated within and may therefore be able to direct cell fate.


Asunto(s)
Resinas Acrílicas/química , Colágeno/química , Hidrogeles/síntesis química , Reactivos de Enlaces Cruzados/química , Humanos , Hidrogeles/efectos adversos , Hidrogeles/química , Fenómenos Mecánicos , Células Madre Mesenquimatosas/efectos de los fármacos , Resistencia a la Tracción
9.
J Biomed Mater Res A ; 103(10): 3188-200, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25777813

RESUMEN

We have used the additive manufacturing technology of selective laser sintering (SLS), together with post SLS heat treatment, to produce porous three dimensional scaffolds from the glass-ceramic apatite-wollastonite (A-W). The A-W scaffolds were custom-designed to incorporate a cylindrical central channel to increase cell penetration and medium flow to the center of the scaffolds under dynamic culture conditions during in vitro testing and subsequent in vivo implantation. The scaffolds were seeded with human bone marrow mesenchymal stromal cells (MSCs) and cultured in spinner flasks. Using confocal and scanning electron microscopy, we demonstrated that MSCs formed and maintained a confluent layer of viable cells on all surfaces of the A-W scaffolds during dynamic culture. MSC-seeded, with and without osteogenic pre-differentiation, and unseeded A-W scaffolds were implanted subcutaneously in MF1 nude mice where osteoid formation and tissue in-growth were observed following histological assessment. The results demonstrate that the in vivo biocompatibility and osteo-supportive capacity of A-W scaffolds can be enhanced by SLS-custom design, without the requirement for osteogenic pre-induction, to advance their potential as patient-specific bone replacement materials.


Asunto(s)
Apatitas/química , Proliferación Celular , Cerámica/química , Ensayo de Materiales , Células Madre Mesenquimatosas/metabolismo , Ácido Silícico/química , Andamios del Tejido/química , Animales , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos
10.
Proc Inst Mech Eng H ; 228(10): 1035-42, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25316684

RESUMEN

Bone remodelling is a vital process which enables bone to repair, renew and optimize itself. Disorders in the bone remodelling process are inevitably manifested in bone-related diseases, such as hypothyroidism, primary hyperparathyroidism and osteoporosis. In our previous work, a predator-prey based mathematical model was developed to simulate bone remodelling cycles under normal and two pathological conditions, hypothyroidism and primary hyperparathyroidism, for trabecular bone at a fixed point. However, the biochemical meanings of the model parameters were not fully explored. This article first extends the previous work by proposing relationships between the model parameters and biochemical factors involved in the bone remodelling process and by examining whether those relationships do predict the behaviours observed in vivo. The model is then applied to the simulation and investigation of bone remodelling of postmenopausal osteoporosis. The proposed connections are supported by good agreement between the model simulations and published experimental observations for the normal condition and all three pathological variations in bone remodelling.


Asunto(s)
Remodelación Ósea/fisiología , Huesos/fisiopatología , Modelos Biológicos , Osteoporosis Posmenopáusica/fisiopatología , Osteoprotegerina/metabolismo , Conducta Predatoria/fisiología , Ligando RANK/metabolismo , Animales , Relojes Biológicos , Simulación por Computador , Femenino , Humanos , Persona de Mediana Edad , Oscilometría/métodos , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal
11.
Biomed Mater Eng ; 24(6): 3373-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25227047

RESUMEN

Multiple myeloma (MM)-induced bone disease is mortal for most MM patients. Bisphosphonates are first-line treatment for MM-induced bone disease, since it can inhibit osteoclast activity and the resultant bone resorption by suppressing the differentiation of osteoclast precursors into mature osteoclasts, promoting osteoclast apoptosis and disrupting osteoclast function. However, it is still unclear whether bisphosphonates have an anti-tumour effect. In our previous work, a computational model was built to simulate the pathology of MM-induced bone disease. This paper extends this proposed computational model to investigate the efficacy of bisphosphonates treatment and then clear the controversy of this therapy. The extended model is validated through the good agreement between simulation results and experimental data. The simulation results suggest that bisphosphonates indeed have an anti-tumour effect.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/fisiopatología , Difosfonatos/administración & dosificación , Quimioterapia Asistida por Computador/métodos , Modelos Biológicos , Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Animales , Conservadores de la Densidad Ósea/administración & dosificación , Resorción Ósea/etiología , Resorción Ósea/patología , Simulación por Computador , Relación Dosis-Respuesta a Droga , Humanos , Mieloma Múltiple/complicaciones , Resultado del Tratamiento
12.
Int J Numer Method Biomed Eng ; 30(11): 1085-102, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24817420

RESUMEN

Multiple myeloma (MM) is the second most common haematological malignancy and results in destructive bone lesions. The interaction between MM cells and the bone microenvironment plays an important role in the development of the tumour cells and MM-induced bone disease and forms a 'vicious cycle' of tumour development and bone destruction, intensified by suppression of osteoblast activity and promotion of osteoclast activity. In this paper, a mathematical model is proposed to simulate how the interaction between MM cells and the bone microenvironment facilitates the development of the tumour cells and the resultant bone destruction. It includes both the roles of inhibited osteoblast activity and stimulated osteoclast activity. The model is able to mimic the temporal variation of bone cell concentrations and resultant bone volume after the invasion and then removal of the tumour cells and explains why MM-induced bone lesions rarely heal even after the complete removal of MM cells. The behaviour of the model compares well with published experimental data. The model serves as a first step to understand the development of MM-induced bone disease and could be applied further to evaluate the current therapies against MM-induced bone disease and even suggests new potential therapeutic targets.


Asunto(s)
Enfermedades Óseas/patología , Modelos Biológicos , Mieloma Múltiple/patología , Algoritmos , Enfermedades Óseas/etiología , Enfermedades Óseas/metabolismo , Humanos , Mieloma Múltiple/complicaciones , Mieloma Múltiple/metabolismo , Osteoblastos/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
Stem Cell Res ; 12(2): 415-27, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24382458

RESUMEN

Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) can differentiate into multiple lineages including osteogenic and adipogenic cells. Wnt signalling has been implicated in controlling BMSC fate, but the mechanisms are unclear and apparently conflicting data exist. Here we show that a novel glycogen synthase kinase 3ß inhibitor, AR28, is a potent activator of canonical Wnt signalling using in vitro ß-catenin translocation studies and TCF-reporter assays. In vivo, AR28 induced characteristic axis duplication and secondary regions of chordin expression in Xenopus laevis embryos. Using human BMSCs grown in adipogenic medium, we confirmed that AR28-mediated Wnt signalling caused a significant (p<0.05) dose-dependent reduction of adipogenic markers. In osteogenic media, including dexamethasone, AR28 caused significant (p<0.05) decreases in alkaline phosphatase (ALP) activity compared to vehicle controls, indicative of a reduced osteogenic response. However, when excluding dexamethasone from the osteogenic media, increases in both ALP and mineralisation were identified following AR28 treatment, which was blocked by mitomycin C. Pre-treatment of BMSCs with AR28 for 7 days before osteogenic induction also increased ALP activity and mineralisation. Furthermore, BMP2-induced osteogenic differentiation was strongly enhanced by AR28 addition within 3 days, but without concomitant changes in cell number, therefore revealing BMP-dependent and independent mechanisms for Wnt-induced osteogenesis.


Asunto(s)
Células de la Médula Ósea/citología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células Madre Mesenquimatosas/citología , Proteínas Wnt/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Dexametasona/farmacología , Sinergismo Farmacológico , Glucógeno Sintasa Quinasa 3 beta , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C3H , Osteogénesis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Xenopus laevis
14.
J Stem Cell Res Ther ; 3(Suppl 6): 004, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24729944

RESUMEN

INTRODUCTION: For clinical applications, Mesenchymal Stromal Cells (MSC) can be isolated from bone marrow and adipose tissue of autologous or allogeneic origin. Allogeneic cell usage has advantages but may harbor the risk of sensitization against foreign HLA. Therefore, we evaluated whether bone marrow and adipose tissue-derived MSC are capable of inducing HLA-specific alloreactivity. METHODS: MSC were isolated from healthy human Bone Marrow (BM-MSC) and adipose tissue (ASC) donors. Peripheral Blood Mononuclear Cells (PBMC) were co-cultured with HLA-AB mismatched BM-MSC or ASC precultured with or without IFNy. After isolation via FACS sorting, the educated CD8+ T effector populations were exposed for 4 hours to Europium labeled MSC of the same HLA make up as in the co-cultures or with different HLA. Lysis of MSC was determined by spectrophotometric measurement of Europium release. RESULTS: CD8+ T cells educated with BM-MSC were capable of HLA specific lysis of BM-MSC. The maximum lysis was 24% in an effector:target (E:T) ratio of 40:1. Exposure to IFNγ increased HLA-I expression on BM-MSC and increased lysis to 48%. Co-culturing of PBMC with IFNγ-stimulated BM-MSC further increased lysis to 76%. Surprisingly, lysis induced by ASC was significantly lower. CD8+ T cells educated with ASC induced a maximum lysis of 13% and CD8+ T cells educated with IFNγ-stimulated ASC of only 31%. CONCLUSION: Allogeneic BM-MSC, and to a lesser extend ASC, are capable of inducing HLA specific reactivity. These results should be taken into consideration when using allogeneic MSC for clinical therapy.

15.
J Stem Cell Res Ther ; Suppl 6(1): 20780, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-24672744

RESUMEN

BACKGROUND: Recent studies with bone marrow (BM)-derived Mesenchymal Stromal Cells (MSC) in transplant recipients demonstrate that treatment with MSC is safe and clinically feasible. While BM is currently the preferred source of MSC, adipose tissue is emerging as an alternative. To develop efficient therapies, there is a need for preclinical efficacy studies in transplantation. We used a unique humanized transplantation model to study the in vivo immunosuppressive effect of human BM-MSC and adipose tissue-derived MSC (ASC). METHODS: Gene expression of BM-MSC and ASC and their capacity to inhibit activated PBMC proliferation was evaluated. The in vivo immunosuppressive effect of BM-MSC and ASC was studied in a humanized mouse model. SCID mice were transplanted with human skin grafts and injected with human allogeneic PBMC with or without administration of BM-MSC or ASC. The effect of MSC on skin graft rejection was studied by immunohistochemistry and PCR. RESULTS: BM-MSC and ASC expressed TGFß, CXCL-10 and IDO. IDO expression and acitivity increased significantly in BM-MSC and ASC upon IFN-γ stimulation. IFN-γ stimulated BM-MSC and ASC inhibited the proliferation of activated PBMC in a significant and dose dependent manner. In our humanized mouse model, alloreactivity was marked by pronounced CD45+ T-cell infiltrates consisting of CD4+ and CD8+ T cells and increased IFN-γ expression in the skin grafts which were all significantly inhibited by both BM-MSC and ASC. CONCLUSION: BM-MSC and ASC are immunosuppressive in vitro and suppress alloreactivity in a preclinical humanized transplantation model.

16.
Methods Mol Biol ; 916: 31-45, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22914931

RESUMEN

In recent years there has been a growing interest in culturing adherent cells using three-dimensional (3D) techniques, rather than more conventional 2D culture methods. This interest emerges from the realization that growing cells on plastic surfaces cannot truly re-create 3D in vivo conditions and therefore might be limiting the cells' potential. In addition, adult stem cells exist in specialized microenvironments, or niches, where the spatial organization of different niche elements (such as different cell types, extracellular matrix) contributes significantly to stem cell maintenance, which cannot be represented using 2D in vitro models. We have generated a range of different 3D approaches for the analysis of mesenchymal stem cells (MSCs) using both mono- and co-culture environments.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Supervivencia Celular , Técnicas de Cocultivo , Criopreservación , Citometría de Flujo , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Hígado/citología , Células Madre Mesenquimatosas/metabolismo , Esferoides Celulares/citología , Coloración y Etiquetado , Telomerasa/genética , Telomerasa/metabolismo
17.
Cytotherapy ; 13(8): 903-12, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21696237

RESUMEN

Mesenchymal stromal cells (MSC) can be isolated from adult tissues and induced to differentiate into skeletal cells, such as osteoblasts, chondrocytes and adipocytes. Consequently, ex vivo MSC are valuable systems for studying the mechanisms that control tissue-context lineage commitment and may offer broad therapeutic applications in the orthopedic theater and beyond. To date, most of these studies have used MSC grown on two-dimensional (2-D) plastic surfaces. The use of three-dimensional (3-D) in vitro growth techniques for MSC may accelerate these areas of research by providing a more representative 'in vivo-like' environment, where cells interact with each other and their cellular products, rather than a plastic surface. We introduce some of the techniques used for 3-D in vitro cultures and how they relate to the MSC field. We will present evidence of how MSC grown as 3-D spheroids not only permits appropriate MSC-like behavior, but appears to promote their stem-cell attributes and therapeutic benefit in applications ranging from regenerative medicine to anti-inflammatory treatments and cancer therapy. 3-D culture techniques also allow de/reconstruction of the specialized in vivo niche of the tissue-resident stem cell where microenvironmental influences can be recognized.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Células Madre Multipotentes/fisiología , Investigación con Células Madre , Animales , Comunicación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Regeneración Tisular Dirigida/tendencias , Humanos , Técnicas de Cultivo de Órganos , Nicho de Células Madre
18.
Stem Cells Dev ; 20(3): 391-403, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20536359

RESUMEN

Emerging data suggest that mesenchymal stem cells (MSCs) are part of a periendothelial niche, suggesting the existence of heterotypic cell-cell crosstalk between endothelial cells and MSCs that regulate MSCs in their local microenvironment. We determined the effects of paracrine factors secreted by human umbilical vein endothelial cells (HUVECs) on MSC survival, proliferation, and differentiation by using an optimized, serum-free HUVEC-conditioned medium (CM). HUVEC-CM induced a significant increase in the size and number of colony-forming units-fibroblast (CFU-F) and CFU-osteoblast (CFU-O) and stimulated the proliferation of MSCs as determined by 5-bromo-2'-deoxyuridine incorporation, compared with non-CM. We also demonstrated that CM significantly enhanced the osteogenic differentiation of MSCs as shown by alkaline phosphatase enzyme histochemistry and von Kossa staining of mineralized nodules as well as by quantitative reverse transcriptase-polymerase chain reaction analysis of osteogenic markers. In contrast, there was no effect on the adipogenic differentiation of MSCs. Bioinformatic integration of HUVEC and MSC gene expression datasets identified several candidate signaling pathways responsible for mediating these effects, including fibroblast growth factor, Wnt, bone morphogenetic protein, and Notch. These data suggest strongly that endothelial cells secrete a soluble factor (or factors) that stimulates progenitor cell activity and, selectively, the osteogenic differentiation of MSCs that could contribute to niche exit.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina , Adipocitos/citología , Adipocitos/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Androstadienos/farmacología , Bencimidazoles/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteocitos/citología , Osteocitos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Cordón Umbilical/citología , Wortmanina
19.
Transplantation ; 90(2): 124-6, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20606604

RESUMEN

There is evolving interest in the use of mesenchymal stem cells (MSC) in solid organ transplantation. Pre-clinical transplantation models show efficacy of MSC in prolonging graft survival and a number of clinical studies are planned or underway. At a recent meeting of the MISOT consortium (MSC In Solid Organ Transplantation) the advances of these studies were evaluated and mechanisms underlying the potential effects of MSC discussed. Continued discussion is required for definition of safety and eventually efficacy endpoints for MSC therapy in solid organ transplantation.


Asunto(s)
Supervivencia de Injerto/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Órganos/métodos , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Inmunofenotipificación , Terapia de Inmunosupresión/métodos , Trasplante de Riñón/inmunología , Trasplante de Riñón/fisiología , Trasplante de Hígado/inmunología , Trasplante de Hígado/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Trasplante de Órganos/fisiología , Seguridad , Linfocitos T/inmunología
20.
J Bone Miner Res ; 25(12): 2680-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20593410

RESUMEN

Mesenchymal stem cell differentiation is controlled by the cooperative activity of a network of signaling mechanisms. Among these, RUNX2 and SOX9 are the major transcription factors for osteogenesis and chondrogenesis, respectively. Their expression is overlapped both temporally and spatially during embryogenesis. Here we have demonstrated that RUNX2 and SOX9 physically interact in intact cells and have confirmed that SOX9 can inhibit the transactivation of RUNX2. In addition, RUNX2 exerts reciprocal inhibition on SOX9 transactivity. In analyses of the mechanism by which SOX9 regulated RUNX2 function, we demonstrated that SOX9 induced a dose-dependent degradation of RUNX2. Although RUNX2 is normally degraded by the ubiquitin-proteasome pathway, we found that SOX9-mediated degradation was proteasome-independent but phosphorylation-dependent and required the presence of the RUNX2 C-terminal domain, which contains a nuclear matrix targeting sequence (NMTS). Furthermore, SOX9 was able to decrease the level of ubiquitinated RUNX2 and direct RUNX2 to the lysosome for degradation. SOX9 also preferentially directed ß-catenin, an intracellular mediator of canonical Wnt signaling, for lysosomal breakdown. Consequently, the mechanisms by which SOX9 regulates RUNX2 function may underlie broader signaling pathways that can influence osteochondrogenesis and mesenchymal fate.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Espacio Intracelular/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Transcripción SOX9/metabolismo , Activación Transcripcional/genética , Animales , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Células HEK293 , Humanos , Lisosomas/metabolismo , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estabilidad Proteica , Ubiquitina/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA