Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 38(9): 1124-1136, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32510174

RESUMEN

Although the application of human mesenchymal stem cells (hMSCs) to repair damaged or diseased tissues has proven relatively effective, both the donor-to-donor variability in ex vivo expansion rates and the maintenance of stemness remain a bottleneck to widespread translation. Previous work from this laboratory stratified donors into those yielding hMSCs with high- or low-growth capacity; global transcriptomic analysis revealed that high-growth-capacity hMSCs were characterized by a loss of the gene encoding glutathione S-transferase theta 1 (GSTT1). These GSTT1-null hMSCs demonstrated increased proliferative rates, clonogenic potential, and longer telomeres compared with low-growth capacity hMSCs that were GSTT1-positive. Thus, this study identifies GSTT1 as a novel genomic DNA biomarker for hMSC scalability.


Asunto(s)
Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Genoma Humano , Células Madre Mesenquimatosas/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Células Clonales , Genotipo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Homocigoto , Humanos , Células Madre Mesenquimatosas/metabolismo , Transcriptoma/genética
2.
Stroke ; 51(9): 2844-2853, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772683

RESUMEN

BACKGROUND AND PURPOSE: Although VEGF165 (vascular endothelial growth factor-165) is able to enhance both angiogenesis and neurogenesis, it also increases vascular permeability through the blood-brain barrier. Heparan sulfate (HS) sugars play important roles in regulating VEGF bioactivity in the pericellular compartment. Here we asked whether an affinity-purified VEGF165-binding HS (HS7) could augment endogenous VEGF activity during stroke recovery without affecting blood-brain barrier function. METHODS: Both rat brain endothelial cell line 4 and primary rat neural progenitor cells were used to evaluate the potential angiogenic and neurogenic effects of HS7 in vitro. For in vivo experiments, male Sprague-Dawley rats were subjected to 100 minutes of transient focal cerebral ischemia, then treated after 4 days with either PBS or HS7. One week later, infarct volume, behavioral sequelae, immunohistochemical markers of angiogenesis and neural stem cell proliferation were assessed. RESULTS: HS7 significantly enhanced VEGF165-mediated angiogenesis in rat brain endothelial cell line 4 brain endothelial cells, and increased the proliferation and differentiation of primary neural progenitor cells, both via the VEGFR2 (vascular endothelial growth factor receptor 2) pathway. Intracerebroventricular injection of HS7 improved neurological outcome in ischemic rats without changing infarct volumes. Immunostaining of the compromised cerebrum demonstrated increases in collagen IV/Ki67 and nestin/Ki67 after HS7 exposure, consistent with its ability to promote angiogenesis and neurogenesis, without compromising blood-brain barrier integrity. CONCLUSIONS: A VEGF-activating glycosaminoglycan sugar, by itself, is able to enhance endogenous VEGF165 activity during the post-ischemic recovery phase of stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Heparitina Sulfato/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/administración & dosificación , Infarto de la Arteria Cerebral Media/prevención & control , Inyecciones Intraventriculares , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/fisiopatología , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Proteomics ; 19(21-22): e1800466, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31197945

RESUMEN

Increasing knowledge of how peptides bind saccharides, and of how saccharides bind peptides, is starting to revolutionize understanding of cell-extracellular matrix relationships. Here, a historical perspective is taken of the relationship between heparan sulfate glycosaminoglycans and how they interact with peptide growth factors in order to both drive and modulate signaling through the appropriate cognate receptors. Such knowledge is guiding the preparation of targeted sugar mimetics that will impact the treatment of many different kinds of diseases, including cancer.


Asunto(s)
Glicómica , Heparitina Sulfato/genética , Péptidos/genética , Proteómica , Matriz Extracelular/genética , Glicosaminoglicanos/genética , Humanos , Neoplasias/genética , Unión Proteica/genética , Transducción de Señal/genética
4.
Angiogenesis ; 21(4): 777-791, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29777314

RESUMEN

Peripheral arterial disease is a major cause of limb loss and its prevalence is increasing worldwide. As most standard-of-care therapies yield only unsatisfactory outcomes, more options are needed. Recent cell- and molecular-based therapies that have aimed to modulate vascular endothelial growth factor-165 (VEGF165) levels have not yet been approved for clinical use due to their uncertain side effects. We have previously reported a heparan sulphate (termed HS7) tuned to avidly bind VEGF165. Here, we investigated the ability of HS7 to promote vascular recovery in a murine hindlimb vascular ischaemia model. HS7 stabilised VEGF165 against thermal and enzyme degradation in vitro, and isolated VEGF165 from serum via affinity-chromatography. C57BL6 mice subjected to unilateral hindlimb ischaemia injury received daily intramuscular injections of respective treatments (n = 8) and were assessed over 3 weeks by laser Doppler perfusion, magnetic resonance angiography, histology and the regain of function. Mice receiving HS7 showed improved blood reperfusion in the footpad by day 7. In addition, they recovered hindlimb blood volume two- to fourfold faster compared to the saline group; the greatest rate of recovery was observed in the first week. Notably, 17% of HS7-treated animals recovered full hindlimb function by day 7, a number that grew to 58% and 100% by days 14 and 21, respectively. This was in contrast to only 38% in the control animals. These results highlight the potential of purified glycosaminoglycan fractions for clinical use following vascular insult, and confirm the importance of harnessing the activity of endogenous pro-healing factors generated at injury sites.


Asunto(s)
Heparitina Sulfato/farmacología , Miembro Posterior , Isquemia/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Miembro Posterior/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isquemia/patología , Isquemia/fisiopatología , Ratones , Células RAW 264.7
5.
J Cell Physiol ; 232(3): 566-575, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27291835

RESUMEN

The future of human mesenchymal stem cells (hMSCs) as a successful cell therapy relies on bioprocessing strategies to improve the scalability of these cells without compromising their therapeutic ability. The culture-expansion of hMSCs can be enhanced by supplementation with growth factors, particularly fibroblast growth factor 2 (FGF2). The biological activity of FGF2 is controlled through interactions with heparan sulfate (HS) that facilitates ligand-receptor complex formation. We previously reported on an FGF2-interacting HS variant (termed HS2) isolated from embryonic tissue by anionic exchange chromatography that increased the proliferation and potency of hMSCs. Here, we detail the isolation of an FGF2 affinity-purified HS variant (HS8) using a scalable platform technology previously employed to generate HS variants with increased affinity for BMP-2 or VEGF165 . This process used a peptide sequence derived from the heparin-binding domain of FGF2 as a substrate to affinity-isolate HS8 from a commercially available source of porcine mucosal HS. Our data show that HS8 binds to FGF2 with higher affinity than to FGF1, FGF7, BMP2, PDGF-BB, or VEGF165 . Also, HS8 protects FGF2 from thermal destabilization and increases FGF signaling and hMSC proliferation through FGF receptor 1. Long-term supplementation of cultures with HS8 increased both hMSC numbers and their colony-forming efficiency without adversely affecting the expression of hMSC-related cell surface antigens. This strategy further exemplifies the utility of affinity-purifying HS variants against particular ligands important to the stem cell microenvironment and advocates for their addition as adjuvants for the culture-expansion of hMSCs destined for cellular therapy. J. Cell. Physiol. 232: 566-575, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Secuencia de Aminoácidos , Anticoagulantes/farmacología , Proliferación Celular , Cromatografía de Afinidad , Disacáridos/análisis , Factor Xa/metabolismo , Factor 2 de Crecimiento de Fibroblastos/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Péptidos/química , Péptidos/metabolismo , Estabilidad Proteica/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Stem Cells ; 33(6): 1878-91, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25752682

RESUMEN

This study sought to identify critical determinants of mesenchymal stem cell (MSC) potency using in vitro and in vivo attributes of cells isolated from the bone marrow of age- and sex-matched donors. Adherence to plastic was not indicative of potency, yet capacity for long-term expansion in vitro varied considerably between donors, allowing the grouping of MSCs from the donors into either those with high-growth capacity or low-growth capacity. Using this grouping strategy, high-growth capacity MSCs were smaller in size, had greater colony-forming efficiency, and had longer telomeres. Cell-surface biomarker analysis revealed that the International Society for Cellular Therapy (ISCT) criteria did not distinguish between high-growth capacity and low-growth capacity MSCs, whereas STRO-1 and platelet-derived growth factor receptor alpha were preferentially expressed on high-growth capacity MSCs. These cells also had the highest mean expression of the mRNA transcripts TWIST-1 and DERMO-1. Irrespective of these differences, both groups of donor MSCs produced similar levels of key growth factors and cytokines involved in tissue regeneration and were capable of multilineage differentiation. However, high-growth capacity MSCs produced approximately double the volume of mineralized tissue compared to low-growth capacity MSCs when assessed for ectopic bone-forming ability. The additional phenotypic criteria presented in this study when combined with the existing ISCT minimum criteria and working proposal will permit an improved assessment of MSC potency and provide a basis for establishing the quality of MSCs prior to their therapeutic application.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Madre Mesenquimatosas/citología , Animales , Células Cultivadas , Citocinas/metabolismo , Humanos , Ratones , Cicatrización de Heridas/fisiología
8.
Glycobiology ; 25(12): 1491-504, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26306634

RESUMEN

Transforming growth factor-ß1 (TGF-ß1, Uniprot: P01137) is a heparin-binding protein that has been implicated in a number of physiological processes, including the initiation of chondrogenesis by human mesenchymal stem cells (hMSCs). Here, we identify the molecular features in the protein and in heparin required for binding and their effects on the potentiation of TGF-ß1's activity on hMSCs. Using a proteomics "Protect and Label" approach, lysines K291, K304, K309, K315, K338, K373, K375 and K388 were identified as being directly involved in binding heparin (Data are available via ProteomeXchange with identifier PXD002772). Competition assays in an optical biosensor demonstrated that TGF-ß1 does require N- and 6-O-sulfate groups for binding but that 2-O-sulfate groups are unlikely to underpin the interaction. Heparin-derived oligosaccharides as short as degree of polymerization (dp) 4 have a weak ability to compete for TGF-ß1 binding to heparin, which increases with the length of the oligosaccharide to reach a maximum between dp18 and dp24. In cell-based assays, heparin, 2-O-, 6-O- and N-desulfated re-N-acetylated heparin and oligosaccharides 14-24 saccharides (dp14-24) in length all increased the phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) after 6 h of stimulation with TGF-ß1. The results provide the structural basis for a model of heparin/heparan sulfate binding to TGF-ß1 and demonstrate that the features in the polysaccharide required for binding are not identical to those required for sustaining the signaling by TGF-ß1 in hMSCs.


Asunto(s)
Heparina/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Células Cultivadas , Heparina/química , Humanos , Células Madre Mesenquimatosas/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
9.
Mol Cancer ; 14: 136, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26201468

RESUMEN

BACKGROUND: Aberrant activation of fibroblast growth factor receptors (FGFRs) deregulates cell proliferation and promotes cell survival, and may predispose to tumorigenesis. Therefore, selective inactivation of FGFRs is an important strategy for cancer therapy. Here as a proof-of-concept study, we developed a FGFR1 neutralizing antisera, IMB-R1, employing a novel strategy aimed at preventing the access of essential heparan sulfate (HS) co-receptors to the heparin-binding domain on FGFR1. METHODS: The mRNA and protein expression level of FGFR1 and other FGFRs were examined in several lines of breast cancer and osteosarcoma cells and corresponding normal cells using Taqman real-time quantitative PCR and Western blot analysis. The specificity of IMB-R1 against FGFR1 was assessed with various ELISA-based approaches and Receptor Tyrosine Kinase array. Proliferation assay and apoptosis analysis were performed to assess the effect of IMB-R1 on cancer cell growth and apoptosis, respectively, in comparison with known FGFR1 inhibitors. The IMB-R1 induced alteration of intracellular signaling and gene expression were analysed using Western blot and microarray approaches. Immunohistochemical staining of FGFR1 using IMB-R1 were carried out in different cancer tissues from clinical patients. Throughout the study, statistical differences were determined by Student's t test where appropriate and reported when a p value was less than 0.05. RESULTS: We demonstrate that IMB-R1 is minimally cross-reactive for other FGFRs, and that it potently and specifically inhibits binding of heparin to FGFR1. Furthermore, IMB-R1 blocks the interaction of FGF2 with FGFR1, the kinase activity of FGFR1 and activation of intracellular FGFR signaling. Cancer cells treated with IMB-R1 displayed impaired FGF2 signaling, were unable to grow and instead underwent apoptosis. IMB-R1-induced cell death correlated with a disruption of antioxidative defense networks and increased expression of several tumor suppressors and apoptotic proteins, including p53. Immunostaining with IMB-R1 was stronger in human cancer tissues in which the FGFR1 gene is amplified. CONCLUSION: Our study suggests that blocking HS interaction with the heparin-binding domains of FGFR1 inhibited cancer cell growth, which can be an attractive strategy to inactivate cancer-related heparin-binding proteins.


Asunto(s)
Antineoplásicos/farmacología , Heparina/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biopsia , Proliferación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos
10.
Biochem Soc Trans ; 42(3): 703-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24849240

RESUMEN

Most research strategies for cartilage tissue engineering use extended culture with complex media loaded with costly GFs (growth factors) to drive tissue assembly and yet they result in the production of cartilage with inferior mechanical and structural properties compared with the natural tissue. Recent evidence suggests that GAGs (glycosaminoglycans) incorporated into tissue engineering scaffolds can sequester and/or activate GFs and thereby more effectively mimic the natural ECM (extracellular matrix). Such approaches may have potential for the improvement of cartilage engineering. However, natural GAGs are structurally complex and heterogeneous, making structure-function relationships hard to determine and clinical translation difficult. Importantly, subfractions of GAGs with specific chain lengths and sulfation patterns have been shown to activate key signalling processes during stem cell differentiation. In addition, recently, GAGs have been bound to synthetic biomaterials, such as electrospun scaffolds and hydrogels, in biologically active conformations, and methods to purify and select affinity-matched GAGs for specific GFs have also been developed. The identification and use of specific GAG moieties to promote chondrogenesis is therefore an exciting new avenue of research. Combining these with synthetic biomaterials may allow a more effective mimicry of the natural ECM, reduction in the need for expensive GFs, and perhaps the deposition of an articular cartilage-like matrix in a clinically relevant manner.


Asunto(s)
Cartílago/fisiología , Linaje de la Célula , Glicosaminoglicanos/fisiología , Regeneración , Cartílago/citología , Cartílago/metabolismo , Glicosaminoglicanos/metabolismo , Humanos
11.
Stem Cells ; 31(12): 2724-36, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23939995

RESUMEN

Signaling through fibroblast growth factor receptor one (FGFR1) is a known inducer of proliferation in both embryonic and human adult mesenchymal stem cells (hMSCs) and positively regulates maintenance of stem cell viability. Leveraging the mitogenic potential of FGF2/FGFR1 signaling in stem cells for therapeutic applications necessitates a mechanistic understanding of how this receptor stimulates cell cycle progression. Using small interfering RNA (siRNA) depletion, antibody-inhibition, and small molecule inhibition, we establish that FGFR1 activity is rate limiting for self-renewal of hMSCs. We show that FGFR1 promotes stem cell proliferation through multiple mechanisms that unite to antagonize cyclin-dependent kinase (CDK) inhibitors. FGFR1 not only stimulates c-Myc to suppress transcription of the CDK inhibitors p21(Waf1) and p27(Kip1), thus promoting cell cycle progression but also increases the activity of protein kinase B (AKT) and the level of S-phase kinase-associated protein 2 (Skp2), resulting in the nuclear exclusion and reduction of p21(Waf1). The in vivo importance of FGFR1 signaling for the control of proliferation in mesenchymal progenitor populations is underscored by defects in ventral mesoderm formation during development upon inhibition of its signaling. Collectively, these studies demonstrate that FGFR1 signaling mediates the continuation of MSC growth and establishes a receptor target for enhancing the expansion of mesenchymal progenitors while maintaining their multilineage potential.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Fase G1/fisiología , Humanos , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Fase S/fisiología , Transducción de Señal , Xenopus laevis
12.
Carbohydr Polym ; 333: 121979, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38494232

RESUMEN

Heparan sulfate (HS) is a glycosaminoglycan (GAG) found throughout nature and is involved in a wide range of functions including modulation of cell signalling via sequestration of growth factors. Current consensus is that the specificity of HS motifs for protein binding are individual for each protein. Given the structural complexity of HS the synthesis of libraries of these compounds to probe this is not trivial. Herein we present the synthesis of an HS decamer, the design of which was undertaken rationally from previously published data for HS binding to the growth factor BMP-2. The biological activity of this HS decamer was assessed in vitro, showing that it had the ability to both bind BMP-2 and increase its thermal stability as well as enhancing the bioactivity of BMP-2 in vitro in C2C12 cells. At the same time no undesired anticoagulant effect was observed. This decamer was then analysed in vivo in a rabbit model where higher bone formation, bone mineral density (BMD) and trabecular thickness were observed over an empty defect or collagen implant alone. This indicated that the HS decamer was effective in promoting bone regeneration in vivo.


Asunto(s)
Glicosaminoglicanos , Heparitina Sulfato , Animales , Conejos , Heparitina Sulfato/química , Osteogénesis , Unión Proteica , Regeneración Ósea , Péptidos y Proteínas de Señalización Intercelular/metabolismo
13.
J Biol Chem ; 286(20): 17755-65, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454472

RESUMEN

We compare here the structural and functional properties of heparan sulfate (HS) chains from both male or female adult mouse liver through a combination of molecular sieving, enzymatic cleavage, and strong anion exchange-HPLC. The results demonstrated that male and female HS chains are significantly different by a number of parameters; size determination showed that HS chain lengths were ∼100 and ∼22 kDa, comprising 30-40 and 6-8 disaccharide repeats, respectively. Enzymatic depolymerization and disaccharide composition analyses also demonstrated significant differences in domain organization and fine structure. N-Unsubstituted glucosamine (ΔHexA-GlcNH(3)(+), ΔHexA-GlcNH(3)(+)(6S), ΔHexA(2S)-GlcNH(3)(+), and N-acetylglucosamine (ΔHexA-GlcNAc) are the predominant disaccharides in male mouse liver HS. However, N-sulfated glucosamine (ΔHexA-GlcNSO(3)) is the predominant disaccharide found in female liver. These structurally different male and female liver HS forms exert differential effects on human mesenchymal cell proliferation and subsequent osteogenic differentiation. The present study demonstrates the potential usefulness of gender-specific liver HS for the manipulation of human mesenchymal cell properties, including expansion, multipotentiality, and subsequent matrix mineralization. Our results suggest that HS chains show both tissue- and gender-specific differences in biochemical composition that directly reflect their biological activity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/farmacología , Hígado/química , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Caracteres Sexuales , Adulto , Animales , Femenino , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos CBA
14.
Gene ; 818: 146199, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35093449

RESUMEN

The multilineage differentiation potential of human mesenchymal stem cells (hMSCs) underpins their clinical utility for tissue regeneration. Control of such cell-fate decisions is tightly regulated by different growth factors/cytokines and their cognate receptors. Fibroblast growth factors (FGFs) are among such factors critical for osteogenesis. However, how FGF receptors (FGFRs) help to orchestrate osteogenic progression remains to be fully elucidated. Here, we studied the protein levels of FGFRs during osteogenesis in human adult bone marrow-derived MSCs and discovered a positive correlation between FGFR2 expression and alkaline phosphatase (ALP) activity, an early marker of osteogenesis. Through RNA interference studies, we confirmed the role of FGFR2 in promoting the osteogenic differentiation of hMSCs. Knockdown of FGFR2 resulted in downregulation of pro-osteogenic genes and upregulation of pro-adipogenic genes and adipogenic commitment. Moreover, under osteogenic induction, FGFR2 knockdown resulted in upregulation of Enhancer of Zeste Homolog 2 (EZH2), an epigenetic enzyme that regulates MSC lineage commitment and suppresses osteogenesis. Lastly, we show that serial-passaged hMSCs have reduced FGFR2 expression and impaired osteogenic potential. Our study suggests that FGFR2 is critical for mediating osteogenic fate by regulating the balance of osteo-adipogenic lineage commitment. Therefore, examining FGFR2 levels during serial-passaging of hMSCs may prove useful for monitoring their multipotency.


Asunto(s)
Linaje de la Célula , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Fosfatasa Alcalina/metabolismo , Proliferación Celular , Células Cultivadas , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Osteogénesis/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
15.
Carbohydr Polym ; 282: 119081, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35123736

RESUMEN

Commercial porcine intestinal mucosal heparan sulfate (HS) is a valuable material for research into its biological functions. As it is usually produced as a side-stream of pharmaceutical heparin manufacture, its chemical composition may vary from batch to batch. We analysed the composition and structure of nine batches of HS from the same manufacturer. Statistical analysis of the disaccharide compositions placed these batches in three categories: group A had high GlcNAc and GlcNS, and low GlcN typical of HS; group B had high GlcN and GlcNS, and low GlcNAc; group C had high di- and trisulfated, and low unsulfated and monosulfated disaccharide repeats. These batches could be placed in the same categories based on their 1H NMR spectra and molecular weights. Anticoagulant and growth factor binding activities of these HS batches did not fit within these same groups but were related to the proportions of more highly sulfated disaccharide repeats.


Asunto(s)
Anticoagulantes/química , Heparitina Sulfato/química , Mucosa Intestinal/química , Animales , Disacáridos/análisis , Factor Xa/química , Péptidos y Proteínas de Señalización Intercelular/química , Porcinos
16.
J Biol Chem ; 285(34): 26233-44, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20547765

RESUMEN

A new strategy has emerged to improve healing of bone defects using exogenous glycosaminoglycans by increasing the effectiveness of bone-anabolic growth factors. Wnt ligands play an important role in bone formation. However, their functional interactions with heparan sulfate/heparin have only been investigated in non-osseous tissues. Our study now shows that the osteogenic activity of Wnt3a is cooperatively stimulated through physical interactions with exogenous heparin. N-Sulfation and to a lesser extent O-sulfation of heparin contribute to the physical binding and optimal co-stimulation of Wnt3a. Wnt3a-heparin signaling synergistically increases osteoblast differentiation with minimal effects on cell proliferation. Thus, heparin selectively reduces the effective dose of Wnt3a needed to elicit osteogenic, but not mitogenic responses. Mechanistically, Wnt3a-heparin signaling strongly activates the phosphoinositide 3-kinase/Akt pathway and requires the bone-related transcription factor RUNX2 to stimulate alkaline phosphatase activity, which parallels canonical beta-catenin signaling. Collectively, our findings establish the osteo-inductive potential of a heparin-mediated Wnt3a-phosphoinositide 3-kinase/Akt-RUNX2 signaling network and suggest that heparan sulfate supplementation may selectively reduce the therapeutic doses of peptide factors required to promote bone formation.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Heparina/farmacología , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Wnt/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Humanos , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Transducción de Señal , Proteína Wnt3 , Proteína Wnt3A
17.
Crit Rev Eukaryot Gene Expr ; 21(1): 1-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21967329

RESUMEN

This review summarizes the emerging strategies that exploit the glycosaminoglycan sugar, heparan sulfate (HS), either as a substitute for, or as a supplement to growth factor (GF) therapy for regenerative medicine. Excluding autograft, the administration of GFs is currently the most effective treatment for critical bone repair and restoration. However, major hurdles in the clinical development of GF therapies include the high cost, the unwanted side effects, and the toxicity associated with the physiological overdosing required to achieve a successful outcome. These drawbacks may be overcome with the application of particular HS fractions that have been optimized to bind, recruit and enhance the biological activity of endogenous GF at the site of injury. Three HS-based treatments are discussed here: first, the single, localized, and sustained delivery of HS as a stand-alone therapeutic agent; then, the inclusion of an HS component within a delivery device so as to stabilize and potentiate the bioactivity of the incorporated GF; and finally, the growing use of HS mimetics, particularly for bone repair.


Asunto(s)
Heparitina Sulfato/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Medicina Regenerativa , Proteína Morfogenética Ósea 2/uso terapéutico , Regeneración Ósea/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Sustitución de Medicamentos , Proteoglicanos de Heparán Sulfato/uso terapéutico , Heparina/farmacología , Heparitina Sulfato/administración & dosificación , Heparitina Sulfato/análogos & derivados , Humanos , Péptidos y Proteínas de Señalización Intercelular/efectos adversos , Cicatrización de Heridas
18.
Pharm Res ; 28(6): 1385-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21184148

RESUMEN

PURPOSE: In order to address cell dose limitations associated with the use of cord blood hematopoietic stem cell (HSC) transplantation, we explored the effect of bone marrow stroma-derived heparan sulfate (HS) on the ex vivo expansion of HSCs. METHODS: Heparan sulfate was isolated and purified from the conditioned media of human bone marrow stromal cells and used for the expansion of cord blood-derived CD34(+) cells in the presence of a cocktail of cytokines. RESULTS: The number of myeloid lineage-committed progenitor cells was increased at low dosage of HS as illustrated by an increase in the total number of colony-forming cells (CFC) and colonies of erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) precursors. Notably, the stroma-derived HS did not alter the growth of CD34(+) HSCs or negatively affect the levels of various HSC phenotypic markers after expansion. CONCLUSIONS: This study shows that HS secreted into solution by stromal cells has the capacity to support hematopoietic cytokines in the maintenance and expansion of HSCs. The incorporation of stroma-derived HS as a reagent may improve the efficacy of cord blood HSC transplantation by enhancing the number of committed cells and accelerating the rate of engraftment.


Asunto(s)
Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Heparitina Sulfato/farmacología , Células de la Médula Ósea/química , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Medios de Cultivo Condicionados , Glicosaminoglicanos/química , Glicosaminoglicanos/aislamiento & purificación , Glicosaminoglicanos/farmacología , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/efectos de los fármacos , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Células Progenitoras de Megacariocitos y Eritrocitos/citología , Células Progenitoras de Megacariocitos y Eritrocitos/efectos de los fármacos , Células Madre , Células del Estroma/química
19.
Nanomedicine ; 7(4): 472-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21215332

RESUMEN

This article reports the method of preparation of gadolinium oxide-doped silica nanoparticles (NPs) whose surface has been functionalized to anchor DNA onto it. The silica NP's surface was modified by 3-aminopropyltrimethoxysilane for DNA to bind electrostatically. Silica NPs with low polydispersity and encapsulating gadolinium oxide were prepared in the aqueous core of the reverse micelles. The average size of these spherical silica NPs doped with gadolinium oxide and dispersed in water is ∼ 50 nm as measured by dynamic light scattering and transmission electron microscopy. The plasmid DNA electrostatically held over NP's surface was firmly immobilized and protected from DNase attack. The gadolinium oxide-doped silica NPs are paramagnetic as observed from the nuclear magnetic resonance (NMR) line-broadening effect on proton spectrum of the surrounding water. In vitro transfection efficiencies of these gadolinium oxide-doped and DNA-conjugated silica NPs in COS-7 and 293T cells were found to be about 75% and 77% respectively of that of 'Polyfect®' as positive control. FROM THE CLINICAL EDITOR: This article reports the method of preparation of gadolinium oxide-doped silica nanoparticles (NPs) whose surface has been functionalized to anchor DNA. These NPs are paramagnetic with in vitro transfection efficiencies in COS-7 and 293T cells of about 75% and 77% compared to 'Polyfect®' as positive control.


Asunto(s)
Medios de Contraste/química , Gadolinio/química , Vectores Genéticos/química , Nanopartículas/química , Dióxido de Silicio/química , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Medios de Contraste/efectos adversos , Gadolinio/efectos adversos , Vectores Genéticos/ultraestructura , Humanos , Espectroscopía de Resonancia Magnética , Microscopía Electrónica de Transmisión , Nanopartículas/efectos adversos , Nanopartículas/ultraestructura , Plásmidos , Transfección
20.
Stem Cell Reports ; 16(4): 694-707, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33636113

RESUMEN

Human mesenchymal stem/stromal cell (hMSC)-based cell therapies are promising for treating a variety of diseases. The unique immunomodulatory properties of hMSCs have extended their therapeutic potential beyond tissue regeneration. However, extensive pre-clinical culture expansion inevitably drives cells toward replicative "aging" and a consequent decline in quality. These "in vitro-aged" hMSCs resemble biologically aged cells, which have been reported to show senescence signatures, diminished immunosuppressive capacity, and weakened regenerative potential as well as pro-inflammatory features. In this review, we have surveyed the literature to explore the intimate relationship between the inflammatory status of hMSCs and their in vitro aging process. We posit that a shift from an anti-inflammatory to a pro-inflammatory phenotype of culture-expanded hMSCs contributes to a deterioration in their therapeutic efficacy. Potential molecular and cellular mechanisms underpinning this phenomenon have been discussed. We have also highlighted studies that leverage these mechanisms to make culture-expanded hMSCs more amenable for clinical use.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Senescencia Celular , Inflamación/patología , Células Madre Mesenquimatosas/patología , Ensayos Clínicos como Asunto , Humanos , Terapia de Inmunosupresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA