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










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2235: 27-35, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33576968

RESUMEN

Pericytes are mural cells closely associated with endothelial cells in capillaries and microvessels. They are precursors of mesenchymal stem/stromal cells that have historically been retrospectively characterized in culture. We established a protocol, described in this chapter, to characterize and isolate pericytes from multiple human organs by flow cytometry and fluorescence-activated cell sorting. This prospective purification of pericytes brings us a step forward in the development of strategies for their use in the clinic.


Asunto(s)
Citometría de Flujo/métodos , Pericitos/citología , Pericitos/trasplante , Capilares/citología , Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Cultivadas , Células Endoteliales/citología , Humanos , Células Madre Mesenquimatosas/citología , Microvasos/citología , Pericitos/metabolismo , Fenotipo
2.
Methods Mol Biol ; 2235: 37-45, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33576969

RESUMEN

Pericytes are found in all vascularized organs and are defined anatomically as perivascular cells that closely surround endothelial cells in capillaries and microvessels and are embedded within the same basement membrane. They have been shown to have diverse physiological and pathological functions including regulation of blood pressure, and tissue regeneration and scarring. Fundamental to understanding the role these cells play in these diverse processes is the ability to accurately identify and localize them in vivo. To do this, we have developed multicolor immunohistochemistry protocols described in this chapter.


Asunto(s)
Inmunohistoquímica/métodos , Pericitos/citología , Pericitos/trasplante , Capilares/citología , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Humanos , Microvasos/citología , Pericitos/metabolismo , Fenotipo
3.
Methods Mol Biol ; 2235: 47-59, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33576970

RESUMEN

We report the use of self-assembled peptide (F2/S) hydrogels and cellular metabolomics to identify a number of innate molecules that are integral to the metabolic processes which drive cellular differentiation of multipotent pericyte stem cells. The culture system relies solely on substrate mechanics to induce differentiation in the absence of traditional differentiation media and therefore is a non-invasive approach to assessing cellular behavior at the molecular level and identifying key metabolites in this process. This novel approach demonstrates that simple metabolites can provide an alternative means to direct stem cell differentiation and that biomaterials can be used to identify them simply and quickly.


Asunto(s)
Metabolómica/métodos , Pericitos/citología , Pericitos/trasplante , Animales , Materiales Biocompatibles/metabolismo , Capilares/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Células Endoteliales/citología , Humanos , Hidrogeles/química , Microvasos/citología , Células Madre Multipotentes/efectos de los fármacos , Péptidos/química , Pericitos/metabolismo , Fenotipo
5.
Cytotherapy ; 22(12): 762-771, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32828673

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) isolated from various tissues are under investigation as cellular therapeutics in a wide range of diseases. It is appreciated that the basic biological functions of MSCs vary depending on tissue source. However, in-depth comparative analyses between MSCs isolated from different tissue sources under Good Manufacturing Practice (GMP) conditions are lacking. Human clinical-grade low-purity islet (LPI) fractions are generated as a byproduct of islet isolation for transplantation. MSC isolates were derived from LPI fractions with the aim of performing a systematic, standardized comparative analysis of these cells with clinically relevant bone marrow-derived MSCs (BM MSCs). METHODS: MSC isolates were derived from LPI fractions and expanded in platelet lysate-supplemented medium or in commercially available xenogeneic-free medium. Doubling rate, phenotype, differentiation potential, gene expression, protein production and immunomodulatory capacity of LPIs were compared with those of BM MSCs. RESULTS: MSCs can be readily derived in vitro from non-transplanted fractions resulting from islet cell processing (i.e., LPI MSCs). LPI MSCs grow stably in serum-free or platelet lysate-supplemented media and demonstrate in vitro self-renewal, as measured by colony-forming unit assay. LPI MSCs express patterns of chemokines and pro-regenerative factors similar to those of BM MSCs and, importantly, are equally able to attract immune cells in vitro and in vivo and suppress T-cell proliferation in vitro. Additionally, LPI MSCs can be expanded to therapeutically relevant doses at low passage under GMP conditions. CONCLUSIONS: LPI MSCs represent an alternative source of GMP MSCs with functions comparable to BM MSCs.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Inmunidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Neovascularización Fisiológica , Páncreas/citología , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Humanos , Inmunomodulación , Interferón gamma/metabolismo , Medicina Regenerativa , Linfocitos T/citología
6.
Am J Sports Med ; 47(4): 991-1000, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29554460

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are increasingly being used in the treatment of a wide variety of sports-related conditions. Despite this enthusiasm, the biological properties of MSCs and their effects on musculoskeletal tissue healing remain poorly understood. MSC-based strategies encompass cell populations with heterogeneous phenotypes isolated from multiple tissues and using different methods. Therefore, comprehensive reporting of the source, preparation methods, and characteristics of MSC strategies is essential to enable interpretation of results. PURPOSE: To perform a systematic review of levels of reporting of key variables in MSC preparation and composition for clinical studies evaluating MSC-based therapies in the treatment of musculoskeletal conditions. STUDY DESIGN: Systematic review. METHODS: A systematic review of the clinical orthopaedic and sports medicine literature from 2002 to 2017 was performed. The following inclusion criteria were used: human clinical trials, published in the English language, involving the administration of MSC-based therapies for orthopaedic or sports medicine applications. In vitro or ex vivo studies, editorials, letters to the editor, and studies relating to cosmetic, neurological, or dental applications were excluded. RESULTS: Of the 1259 studies identified on the initial search, 36 studies were found to satisfy the inclusion criteria for analysis on comprehensive review. Fifty-seven percent of studies evaluated bone marrow-derived MSCs, 41% evaluated adipose-derived MSCs, and 2% evaluated synovium-derived MSCs. Considerable deficiencies in the reporting of key variables, including the details of stem cell processing, culture conditions, and the characteristics of cell populations delivered, were noted. Overall, studies reported only 52% (range, 30%-80%) of variables that may critically influence outcome. No study provided adequate information relating to all of these variables. CONCLUSION: All existing clinical studies evaluating MSCs for orthopaedic or sports medicine applications are limited by inadequate reporting of both preparation protocols and composition. Deficient reporting of the variables that may critically influence outcome precludes interpretation, prevents others from reproducing experimental conditions, and makes comparisons across studies difficult. We encourage the adoption of emerging minimum reporting standards for clinical studies evaluating the use of MSCs in orthopaedics.


Asunto(s)
Técnicas de Preparación Histocitológica , Células Madre Mesenquimatosas , Procedimientos Ortopédicos , Traumatismos en Atletas/terapia , Humanos , Trasplante de Células Madre Mesenquimatosas , Enfermedades Musculoesqueléticas/terapia , Sistema Musculoesquelético/lesiones
7.
Arthroscopy ; 34(4): 1366-1375, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29395555

RESUMEN

PURPOSE: To perform a systematic review of clinical studies evaluating bone marrow aspirate concentrate (BMAC) in the treatment of musculoskeletal pathology to compare levels of reporting with recently published minimum standards. METHODS: A systematic review of the clinical literature from August 2002 to August 2017 was performed. Human clinical studies published in English and involving the administration of BMAC for musculoskeletal applications were included. Studies evaluating non-concentrated preparations of bone marrow aspirate or preparations of laboratory cultured cells were excluded. Studies evaluating the treatment of dental or maxillofacial conditions were excluded. Similarly, in vitro studies, editorials, letters to the editor, and reviews were excluded. Levels of reporting were compared with previously published minimum standards agreed on through an international Delphi consensus process. RESULTS: Of 1,580 studies identified on the initial search, 46 satisfied the criteria for inclusion. Considerable deficiencies in reporting of key variables including the details of BMAC preparation and composition were noted. Studies reported information on only 42% (range, 25%-60%) of the variables included within established minimum reporting standards. No study provided adequate information to enable the precise replication of preparation protocols and accurate characterization of the BMAC formulation delivered. CONCLUSIONS: We found that all existing clinical studies in the literature evaluating BMAC for orthopaedic or sports medicine applications are limited by inadequate reporting of both preparation protocols and composition. Deficient reporting of the variables that may critically influence outcomes precludes interpretation, prevents other researchers from reproducing experimental conditions, and makes comparisons across studies difficult. We encourage the adoption of emerging minimum reporting standards for clinical studies evaluating the use of mesenchymal stem cells in orthopaedics. LEVEL OF EVIDENCE: Level IV, systematic review of Level I through IV studies.


Asunto(s)
Trasplante de Médula Ósea , Protocolos Clínicos/normas , Trasplante de Células Madre Mesenquimatosas , Enfermedades Musculoesqueléticas/terapia , Procedimientos Ortopédicos/normas , Humanos
8.
J Burn Care Res ; 39(4): 634-635, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28661979

RESUMEN

This case report describes a gastric acid burn. Gastric acid burns secondary to vomiting in an elderly patient who has fallen and experienced a so called "long lie" have not been reported. The patient was admitted under the physicians and referred to the burns team once the wounds were recognized as burns. The patient was treated nonsurgically with dressings and healed well with conservative treatment. Here, the authors highlight the importance of recognizing and treating burns of this nature in our ageing population.


Asunto(s)
Quemaduras Químicas/etiología , Quemaduras Químicas/terapia , Ácido Gástrico/química , Vómitos/complicaciones , Accidentes por Caídas , Anciano , Traumatismos de la Espalda/inducido químicamente , Tratamiento Conservador , Femenino , Humanos , Traumatismos del Cuello/inducido químicamente , Lesiones del Hombro/inducido químicamente
9.
ACS Biomater Sci Eng ; 3(8): 1710-1718, 2017 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-28824958

RESUMEN

Sandwichlike (SW) cultures are engineered as a multilayer technology to simultaneously stimulate dorsal and ventral cell receptors, seeking to mimic cell adhesion in three-dimensional (3D) environments in a reductionist manner. The effect of this environment on cell differentiation was investigated for several cell types cultured in standard growth media, which promotes proliferation on two-dimensional (2D) surfaces and avoids any preferential differentiation. First, murine C2C12 myoblasts showed specific myogenic differentiation. Human mesenchymal stem cells (hMSCs) of adipose and bone marrow origin, which can differentiate toward a wider variety of lineages, showed again myodifferentiation. Overall, this study shows myogenic differentiation in normal growth media for several cell types under SW conditions, avoiding the use of growth factors and cytokines, i.e., solely by culturing cells within the SW environment. Mechanistically, it provides further insights into the balance between integrin adhesion to the dorsal substrate and the confinement imposed by the SW system.

10.
Stem Cells ; 35(11): 2280-2291, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28833807

RESUMEN

Multipotent mesenchymal stem cells (MSCs) have enormous potential in tissue engineering and regenerative medicine. However, until now, their development for clinical use has been severely limited as they are a mixed population of cells with varying capacities for lineage differentiation and tissue formation. Here, we identify receptor tyrosine kinase-like orphan receptor 2 (ROR2) as a cell surface marker expressed by those MSCs with an enhanced capacity for cartilage formation. We generated clonal human MSC populations with varying capacities for chondrogenesis. ROR2 was identified through screening for upregulated genes in the most chondrogenic clones. When isolated from uncloned populations, ROR2+ve MSCs were significantly more chondrogenic than either ROR2-ve or unfractionated MSCs. In a sheep cartilage-repair model, they produced significantly more defect filling with no loss of cartilage quality compared with controls. ROR2+ve MSCs/perivascular cells were present in developing human cartilage, adult bone marrow, and adipose tissue. Their frequency in bone marrow was significantly lower in patients with osteoarthritis (OA) than in controls. However, after isolation of these cells and their initial expansion in vitro, there was greater ROR2 expression in the population derived from OA patients compared with controls. Furthermore, osteoarthritis-derived MSCs were better able to form cartilage than MSCs from control patients in a tissue engineering assay. We conclude that MSCs expressing high levels of ROR2 provide a defined population capable of predictably enhanced cartilage production. Stem Cells 2017;35:2280-2291.


Asunto(s)
Condrogénesis/genética , Células Madre Mesenquimatosas/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Proteína Wnt-5a/genética , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Ovinos , Ingeniería de Tejidos , Proteína Wnt-5a/metabolismo
11.
Sci Rep ; 7(1): 6895, 2017 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-28761049

RESUMEN

Differentiation of stem cells to chondrocytes in vitro usually results in a heterogeneous phenotype. This is evident in the often detected over expression of type X collagen which, in hyaline cartilage structure is not characteristic of the mid-zone but of the deep-zone ossifying tissue. Methods to better match cartilage developed in vitro to characteristic in vivo features are therefore highly desirable in regenerative medicine. This study compares phenotype characteristics between pericytes, obtained from human adipose tissue, differentiated using diphenylalanine/serine (F2/S) peptide hydrogels with the more widely used chemical induced method for chondrogenesis. Significantly higher levels of type II collagen were noted when pericytes undergo chondrogenesis in the hydrogel in the absence of induction media. There is also a balanced expression of collagen relative to aggrecan production, a feature which was biased toward collagen production when cells were cultured with induction media. Lastly, metabolic profiles of each system show considerable overlap between both differentiation methods but subtle differences which potentially give rise to their resultant phenotype can be ascertained. The study highlights how material and chemical alterations in the cellular microenvironment have wide ranging effects on resultant tissue type.


Asunto(s)
Tejido Adiposo/citología , Condrocitos/citología , Péptidos/química , Pericitos/citología , Tejido Adiposo/metabolismo , Biomarcadores , Diferenciación Celular , Células Cultivadas , Condrocitos/metabolismo , Condrogénesis , Colágeno Tipo II/metabolismo , Dipéptidos , Humanos , Hidrogeles/química , Metabolómica , Pericitos/metabolismo , Fenotipo , Fenilalanina/análogos & derivados , Fenilalanina/química , Ingeniería de Tejidos/métodos
12.
Pharmacol Ther ; 171: 93-103, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27510330

RESUMEN

Pericytes and other perivascular stem cells are of growing interest in orthopedics and tissue engineering. Long regarded as simple regulators of angiogenesis and blood pressure, pericytes are now recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Pericytes are typified by characteristic cell surface marker expression (including αSMA, CD146, PDGFRß, NG2, RGS5, among others). Although alone no marker is absolutely specific for pericytes, collectively these markers appear to selectively identify an MSC-like pericyte. The purification of pericytes is most well described as a CD146+CD34-CD45- cell population. Pericytes and other perivascular stem cell populations have been applied in diverse orthopedic applications, including both ectopic and orthotopic models. Application of purified cells has sped calvarial repair, induced spine fusion, and prevented fibrous non-union in rodent models. Pericytes induce these effects via both direct and indirect mechanisms. In terms of their paracrine effects, pericytes are known to produce and secrete high levels of a number of growth and differentiation factors both in vitro and after transplantation. The following review will cover existing studies to date regarding pericyte application for bone and cartilage engineering. In addition, further questions in the field will be pondered, including the phenotypic and functional overlap between pericytes and culture-derived MSC, and the concept of pericytes as efficient producers of differentiation factors to speed tissue repair.


Asunto(s)
Enfermedades Musculoesqueléticas/terapia , Pericitos/citología , Ingeniería de Tejidos/métodos , Animales , Huesos/citología , Cartílago/citología , Diferenciación Celular , Humanos , Células Madre Mesenquimatosas/citología , Enfermedades Musculoesqueléticas/patología , Roedores , Células Madre/citología
14.
Diabetes ; 64(3): 733-45, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25377876

RESUMEN

Prolyl hydroxylase enzymes (PHDs) sense cellular oxygen upstream of hypoxia-inducible factor (HIF) signaling, leading to HIF degradation in normoxic conditions. In this study, we demonstrate that adipose PHD2 inhibition plays a key role in the suppression of adipocyte lipolysis. Adipose Phd2 gene ablation in mice enhanced adiposity, with a parallel increase in adipose vascularization associated with reduced circulating nonesterified fatty acid levels and normal glucose homeostasis. Phd2 gene-depleted adipocytes exhibited lower basal lipolysis in normoxia and reduced ß-adrenergic-stimulated lipolysis in both normoxia and hypoxia. A selective PHD inhibitor suppressed lipolysis in murine and human adipocytes in vitro and in vivo in mice. PHD2 genetic ablation and pharmacological inhibition attenuated protein levels of the key lipolytic effectors hormone-sensitive lipase and adipose triglyceride lipase (ATGL), suggesting a link between adipocyte oxygen sensing and fatty acid release. PHD2 mRNA levels correlated positively with mRNA levels of AB-hydrolase domain containing-5, an activator of ATGL, and negatively with mRNA levels of lipid droplet proteins, perilipin, and TIP47 in human subcutaneous adipose tissue. Therapeutic pseudohypoxia caused by PHD2 inhibition in adipocytes blunts lipolysis and promotes benign adipose tissue expansion and may have therapeutic applications in obesity or lipodystrophy.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/citología , Lipólisis/fisiología , Tejido Adiposo/metabolismo , Adulto , Animales , Ácidos Grasos no Esterificados/metabolismo , Femenino , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Immunoblotting , Inmunohistoquímica , Lipasa/metabolismo , Masculino , Ratones , Persona de Mediana Edad
15.
Cell Mol Life Sci ; 71(8): 1353-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24158496

RESUMEN

Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.


Asunto(s)
Biomarcadores/metabolismo , Vasos Sanguíneos/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/clasificación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Pericitos/citología , Técnicas de Cultivo de Célula , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación
16.
J Clin Endocrinol Metab ; 99(1): E160-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24243637

RESUMEN

CONTEXT: Levels of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), which regenerates active glucocorticoids, are selectively elevated in adipose tissue in human obesity and metabolic syndrome, both conditions associated with chronic low-grade inflammation. 11ß-HSD1 expression is induced by proinflammatory cytokines in a variety of cell types, including in human adipocytes differentiated in vitro. OBJECTIVE: Our objective was to determine the mechanisms by which proinflammatory cytokines induce 11ß-HSD1 in human adipocytes. RESULTS: The proinflammatory cytokines IL-1α (10 ng/mL) and TNFα (20 ng/mL) increased 11ß-HSD1 mRNA levels in human primary adipocyte fractions and Simpson-Golabi-Behmel syndrome (SGBS) adipocytes (P<.001). Inhibition of the MAPK/ERK kinase (MEK) attenuated CCAAT/enhancer binding protein (C/EBP) ß phosphorylation at Thr235 and IL-1α/TNFα induction of 11ß-HSD1 (P≤.007). The small interfering RNA-mediated knockdown of C/EBPß and nuclear factor (NF)-κB/RelA or inhibition of NF-κB/RelA also attenuated cytokine induction of 11ß-HSD1 (P≤.001). Moreover, induction of 11ß-HSD1 by IL-1α in SGBS cells was associated with nuclear localization of C/EBPß and NF-κB/RelA. Chromatin immunoprecipitation experiments showed C/EBPß and NF-κB/RelA located to the 11ß-HSD1 promoter in human adipose tissue. Treatment of adipocyte fractions or SGBS adipocytes with metformin or acetylsalicylic acid, which target C/EBPß and NF-κB/RelA signaling, attenuated the IL-1α induction of 11ß-HSD1 (P≤.002). CONCLUSIONS: Increased proinflammatory signaling in inflamed adipose tissue may mediate elevated 11ß-HSD1 expression at this site via MEK, C/EBPß, and NF-κB/RelA. These molecules/signaling pathways are, therefore, potential targets for drugs, including metformin and acetylsalicylic acid, to prevent/decreased up-regulation of 11ß-HSD1 in human obese/metabolic syndrome adipose tissue.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/biosíntesis , Adipocitos/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Mediadores de Inflamación/farmacología , FN-kappa B/fisiología , Adipocitos/metabolismo , Adulto , Células Cultivadas , Citocinas/farmacología , Inducción Enzimática/efectos de los fármacos , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Factor de Transcripción ReIA/fisiología , Adulto Joven
17.
Biomater Sci ; 2(11): 1683-1692, 2014 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-32481948

RESUMEN

Mesenchymal stem cells (MSCs) hold great promise in regenerative medicine due to their wide multilineage potential as well as their ability to suppress/modulate the immune response. Maintaining these cells in vitro and expanding them on a clinically relevant scale remains a challenge that needs to be addressed to realise their full potential. Current culture methods for MSCs typically rely on animal sourced substrates and often result in a heterogeneous population of cells with varying degrees of differentiation capacity. Here, a high-throughput platform was used to identify synthetic substrates for MSC culture that not only facilitated growth but also maintained the MSC phenotype. Two polymers, PU157 (synthesised from poly(butyleneglycol) and 4,4'-methylenediphenyldiisocyanate with 3-(dimethylamino)-1,2-propanediol as a chain extender) and PA338 (N-methylaniline modified poly(methylmethacrylate-co-glycidylmethacrylate)) were able to maintain the growth and phenotype of human embryonic derived mesenchymal progenitors (hES-MPs) and adipose derived MSCs (ADMSCs) for five and ten passages, respectively. Cell phenotype and multipotency were confirmed by flow cytometry analysis of ten MSC markers and differentiation analysis. These new polymer substrates provide a chemically defined synthetic surface for efficient, long-term MSC culture.

18.
Cytometry A ; 83(8): 714-20, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23818229

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are adult multipotent progenitors of great promise for cell therapy. MSCs can mediate tissue regeneration, immunomodulation, and hematopoiesis support. Despite the unique properties of MSCs and their broad range of potential clinical applications, the very nature of these cells has been uncertain. Furthermore, MSCs are heterogeneous and only defined subpopulations of these are endowed with the particular abilities to sustain hematopoietic stem cells, regulate immune responses, or differentiate into mesodermal cell lineages. It is becoming evident that current criteria used to define cultured polyclonal MSCs (expression of nonspecific markers and in vitro mesodermal differentiation) are not sufficient to fully understand and exploit the potential of these cells. Here, we describe how flow cytometry has been used to reveal a perivascular origin of MSCs. As a result, the prospective purification of MSCs and specialized subsets thereof is now possible, and the clinical use of purified autologous MSCs is now within reach.


Asunto(s)
Citometría de Flujo , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo Blanco/citología , Animales , Antígenos CD/metabolismo , Vasos Sanguíneos/citología , Separación Celular , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Medicina Regenerativa
19.
J Vis Exp ; (56): e2969, 2011 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22064456

RESUMEN

Despite progress in modelling human drug toxicity, many compounds fail during clinical trials due to unpredicted side effects. The cost of clinical studies are substantial, therefore it is essential that more predictive toxicology screens are developed and deployed early on in drug development (Greenhough et al 2010). Human hepatocytes represent the current gold standard model for evaluating drug toxicity, but are a limited resource that exhibit variable function. Therefore, the use of immortalised cell lines and animal tissue models are routinely employed due to their abundance. While both sources are informative, they are limited by poor function, species variability and/or instability in culture (Dalgetty et al 2009). Pluripotent stem cells (PSCs) are an attractive alternative source of human hepatocyte like cells (HLCs) (Medine et al 2010). PSCs are capable of self renewal and differentiation to all somatic cell types found in the adult and thereby represent a potentially inexhaustible source of differentiated cells. We have developed a procedure that is simple, highly efficient, amenable to automation and yields functional human HLCs (Hay et al 2008 ; Fletcher et al 2008 ; Hannoun et al 2010 ; Payne et al 2011 and Hay et al 2011). We believe our technology will lead to the scalable production of HLCs for drug discovery, disease modeling, the construction of extra-corporeal devices and possibly cell based transplantation therapies.


Asunto(s)
Técnicas Citológicas/métodos , Células Madre Embrionarias/citología , Hepatocitos/citología , Células Madre Pluripotentes/citología , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...