Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Small ; 17(13): e2007551, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33690981

RESUMEN

Biointerface engineering is a wide-spread strategy to improve the healing process and subsequent tissue integration of biomaterials. Especially the integration of specific peptides is one promising strategy to promote the regenerative capacity of implants and 3D scaffolds. In vivo, these tailored interfaces are, however, first confronted with the innate immune response. Neutrophils are cells with pronounced proteolytic potential and the first recruited immune cells at the implant site; nonetheless, they have so far been underappreciated in the design of biomaterial interfaces. Herein, an in vitro approach is introduced to model and analyze the neutrophil interaction with bioactivated materials at the example of nano-bioinspired electrospun surfaces that reveals the vulnerability of a given biointerface design to the contact with neutrophils. A sacrificial, transient hydrogel coating that demonstrates optimal protection for peptide-modified surfaces and thus alleviates the immediate cleavage by neutrophil elastase is further introduced.


Asunto(s)
Materiales Biocompatibles , Elastasa de Leucocito , Humanos , Hidrogeles , Inmunidad Innata , Neutrófilos
2.
J Cell Biochem ; 118(2): 263-275, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27305863

RESUMEN

Controlling the adipo-osteogenic lineage decision of trabecular human bone marrow stromal cells (hBMSCs) in favor of osteogenesis represents a promising approach for osteoporosis therapy and prevention. Previously, Fibroblast Growth Factor 1 (FGF1) and its subfamily member FGF2 were scored as leading candidates to exercise control over skeletal precursor commitment and lineage decision albeit literature results are highly inconsistent. We show here that FGF1 and 2 strongly prevent the osteogenic commitment and differentiation of hBMSCs. Mineralization of extracellular matrix (ECM) and mRNA expression of osteogenic marker genes Alkaline Phosphatase (ALP), Collagen 1A1 (COL1A1), and Integrin-Binding Sialoprotein (IBSP) were significantly reduced. Furthermore, master regulators of osteogenic commitment like Runt-Related Transcription Factor 2 (RUNX2) and Bone Morphogenetic Protein 4 (BMP4) were downregulated. When administered under adipogenic culture conditions, canonical FGFs did not support osteogenic marker expression. Moreover despite the presence of osteogenic differentiation factors, FGFs even disabled the pro-osteogenic lineage decision of pre-differentiated adipocytic cells. In contrast to FGF Receptor 2 (FGFR2), FGFR1 was stably expressed throughout osteogenic and adipogenic differentiation and FGF addition. Moreover, FGFR1 and Extracellular Signal-Regulated Kinases 1 and 2 (ERK1/2) were found to be responsible for underlying signal transduction using respective inhibitors. Taken together, we present new findings indicating that canonical FGFR-ERK1/2 signaling entrapped hBMSCs in a pre-committed state and arrested further maturation of committed precursors. Our results might aid in unraveling and controlling check points relevant for ageing-associated aberrant adipogenesis with consequences for the treatment of degenerative diseases such as osteoporosis and for skeletal tissue engineering strategies. J. Cell. Biochem. 118: 263-275, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Células de la Médula Ósea/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Osteogénesis/efectos de los fármacos , Adulto , Anciano , Antígenos de Diferenciación/biosíntesis , Células de la Médula Ósea/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células del Estroma/citología , Células del Estroma/metabolismo
3.
Exp Cell Res ; 338(2): 136-48, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26384550

RESUMEN

Multipotent human bone marrow stromal cells (hBMSCs) are the common progenitors of osteoblasts and adipocytes. A shift in hBMSC differentiation in favor of adipogenesis may contribute to the bone loss and marrow fat accumulation observed in aging and osteoporosis. Hence, the identification of factors modulating marrow adipogenesis is of great therapeutic interest. Fibroblast growth factors 1 (FGF1) and 2 (FGF2) play important roles in several cellular processes including differentiation. Their role in adipogenesis is, however, still unclear given the contradictory reports found in the literature. In this work, we investigated the effect of FGF signaling on hBMSC adipogenesis in a 3D collagen gel system to mimic the natural microenvironment. We successfully established adipogenic differentiation of hBMSC embedded in type I collagen gels. We found that exogenous FGF1 and FGF2 exerted an inhibitory effect on lipid droplet accumulation and gene expression of adipogenic markers, which was abolished by pharmacological blocking of FGF receptor (FGFR) signaling. FGF treatment also affected the expression of the matrix metalloproteinase 13 (MMP13) and the tissue inhibitor of metalloproteinases 1 (TIMP1), altering the MMP/TIMP balance, which modulates collagen processing and turnover. FGF1- and FGF2-mediated inhibition of differentiation was, however, not restricted to adipogenesis since FGF1 and FGF2 treatment also resulted in the inhibition of the osteogenic differentiation in collagen gels. We conclude that FGFR signaling inhibits the in vitro adipogenic commitment of hBMSCs, downregulating core differentiation markers and altering ECM composition.


Asunto(s)
Adipogénesis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Colágeno Tipo I/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Geles/metabolismo , Células del Estroma/efectos de los fármacos , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 13 de la Matriz/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Células del Estroma/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
4.
Biofabrication ; 12(2): 025007, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31805543

RESUMEN

Macrophages are key players of the innate immune system that can roughly be divided into the pro-inflammatory M1 type and the anti-inflammatory, pro-healing M2 type. While a transient initial pro-inflammatory state is helpful, a prolonged inflammation deteriorates a proper healing and subsequent regeneration. One promising strategy to drive macrophage polarization by biomaterials is precise control over biomaterial geometry. For regenerative approaches, it is of particular interest to identify geometrical parameters that direct human macrophage polarization. For this purpose, we advanced melt electrowriting (MEW) towards the fabrication of fibrous scaffolds with box-shaped pores and precise inter-fiber spacing from 100 µm down to only 40 µm. These scaffolds facilitate primary human macrophage elongation accompanied by differentiation towards the M2 type, which was most pronounced for the smallest pore size of 40 µm. These new findings can be important in helping to design new biomaterials with an enhanced positive impact on tissue regeneration.


Asunto(s)
Macrófagos/citología , Ingeniería de Tejidos/instrumentación , Andamios del Tejido/química , Materiales Biocompatibles/química , Diferenciación Celular , Polaridad Celular , Humanos , Macrófagos/inmunología , Porosidad
5.
Sci Rep ; 9(1): 3533, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837625

RESUMEN

In vitro co-cultures of different primary human cell types are pivotal for the testing and evaluation of biomaterials under conditions that are closer to the human in vivo situation. Especially co-cultures of macrophages and mesenchymal stem cells (MSCs) are of interest, as they are both present and involved in tissue regeneration and inflammatory reactions and play crucial roles in the immediate inflammatory reactions and the onset of regenerative processes, thus reflecting the decisive early phase of biomaterial contact with the host. A co-culture system of these cell types might thus allow for the assessment of the biocompatibility of biomaterials. The establishment of such a co-culture is challenging due to the different in vitro cell culture conditions. For human macrophages, medium is usually supplemented with human serum (hS), whereas hMSC culture is mostly performed using fetal calf serum (FCS), and these conditions are disadvantageous for the respective other cell type. We demonstrate that human platelet lysate (hPL) can replace hS in macrophage cultivation and appears to be the best option for co-cultivation of human macrophages with hMSCs. In contrast to FCS and hS, hPL maintained the phenotype of both cell types, comparable to that of their respective standard culture serum, as well as the percentage of each cell population. Moreover, the expression profile and phagocytosis activity of macrophages was similar to hS.


Asunto(s)
Plaquetas/química , Medios de Cultivo/química , Suero/química , Animales , Plaquetas/metabolismo , Bovinos , Técnicas de Cocultivo , Medios de Cultivo/farmacología , Citocinas/metabolismo , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Fagocitosis/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
6.
ACS Biomater Sci Eng ; 5(12): 6655-6666, 2019 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33423484

RESUMEN

Melt electrowriting (MEW) is an additive manufacturing technology that produces readily handleable fibrous scaffolds with controlled geometry to support cell infiltration. Although MEW scaffolds have excellent potential for cell delivery in regenerative medicine applications, studies to date have primarily focused on polymers such as poly(ε-caprolactone) (PCL) that lack bioactive cues to affect cell function. To address this aspect, MEW scaffolds with extracellular matrix (ECM) coatings were developed as a proadipogenic platform for human mesenchymal stromal cells (hMSCs). More specifically, highly flexible PCL scaffolds fabricated through MEW were coated with a complex ECM suspension prepared from human decellularized adipose tissue (DAT), purified fibronectin, or laminin to determine the effects of two key bioactive proteins present within adipose-derived ECM. In vitro studies exploring the response of human bone marrow-derived mesenchymal stromal cells cultured under adipogenic differentiation conditions indicated a high level of differentiation on all substrates studied, including unmodified PCL scaffolds and two-dimensional controls. To more fully assess the intrinsic proadipogenic capacity of the composite biomaterials, a modified culture regime was established that involved a short-term adipogenic induction in differentiation medium, followed by continued culture in maintenance medium supplemented with insulin for up to 3 weeks. Under these conditions, adipogenic differentiation was enhanced on all fiber scaffolds as compared to the tissue culture controls. Notably, the highest adipogenic response was consistently observed on the PCL + DAT scaffolds, based on the analysis of multiple markers including adipogenic gene [lipoprotein lipase, fatty acid binding protein 4 (FABP4), adiponectin, perilipin 1] and protein (FABP4, leptin) expression and intracellular triglyceride accumulation. Taken together, the PCL scaffolds incorporating DAT provide an adipoinductive microenvironment for the hMSCs, with particular applicability of this cell-instructive delivery platform for applications in plastic and reconstructive surgery.

7.
J Cell Biochem ; 103(2): 413-33, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17610236

RESUMEN

The molecular events associated with the age-related gain of fatty tissue in human bone marrow are still largely unknown. Besides enhanced adipogenic differentiation of mesenchymal stem cells (MSCs), transdifferentiation of osteoblast progenitors may contribute to bone-related diseases like osteopenia. Transdifferentiation of MSC-derived osteoblast progenitors into adipocytes and vice versa has previously been proven feasible in our cell culture system. Here, we focus on mRNA species that are regulated during transdifferentiation and represent possible control factors for the initiation of transdifferentiation. Microarray analyses comparing transdifferentiated cells with normally differentiated cells exhibited large numbers of reproducibly regulated genes for both, adipogenic and osteogenic transdifferentiation. To evaluate the relevance of individual genes, we designed a scoring scheme to rank genes according to reproducibility, regulation level, and reciprocity between the different transdifferentiation directions. Thereby, members of several signaling pathways like FGF, IGF, and Wnt signaling showed explicitly differential expression patterns. Additional bioinformatic analysis of microarray analyses allowed us to identify potential key factors associated with transdifferentiation of adipocytes and osteoblasts, respectively. Fibroblast growth factor 1 (FGF1) was scored as one of several lead candidate gene products to modulate the transdifferentiation process and is shown here to exert inhibitory effects on adipogenic commitment and differentiation.


Asunto(s)
Transdiferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Células Madre Mesenquimatosas/metabolismo , Adipocitos/citología , Adipogénesis/genética , Adulto , Anciano , Envejecimiento/patología , Células de la Médula Ósea/citología , Células Cultivadas/metabolismo , Femenino , Factor 1 de Crecimiento de Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/citología , Osteogénesis/genética , ARN Mensajero/biosíntesis , Transducción de Señal
8.
Mol Cell Endocrinol ; 271(1-2): 1-17, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475397

RESUMEN

We established a cell culture system of human mesenchymal stem cells that allows not only for osteogenic and adipogenic differentiation but also for transdifferentiation between both cell lineages. Committed osteoblasts were transdifferentiated into adipocytes with losing osteogenic but highly expressing adipogenic markers. Adipocytes were transdifferentiated into osteoblasts with most of the resulting cells showing osteogenic but some still displaying adipogenic markers apparently not responding to the reprogramming stimulus. Comparing transdifferentiated adipocytes with committed osteoblasts by microarray analysis revealed 258 regulated transcripts, many of them associated with signal transduction, metabolism, and transcription but mostly distinct from established inducing factors of normal adipogenic and osteogenic differentiation, respectively. The regulation pattern of 20 of 22 selected genes was confirmed by semiquantitative RT-PCR. Our results indicate that the plasticity between osteogenesis and adipogenesis extends into the differentiation pathways of both cell lineages and may contribute to the age-related expansion of adipose tissue in human bone marrow.


Asunto(s)
Adipogénesis/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Osteogénesis/fisiología , Fosfatasa Alcalina/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos
9.
Bone ; 78: 102-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25959412

RESUMEN

Heparins are broadly used for the prevention and treatment of thrombosis and embolism. Yet, osteoporosis is considered to be a severe side effect in up to one third of all patients on long-term treatment. However, the mechanisms underlying this clinical problem are only partially understood. To investigate if heparin affects differentiation of skeletal precursors, we examined the effects of heparin on the osteogenic and adipogenic lineage commitment and differentiation of primary human bone marrow stromal cells (hBMSCs). Due to the known inverse relationship between adipogenesis and osteogenesis and the capacity of pre-differentiated cells to convert into the respective other lineage, we also determined heparin effects on osteogenic conversion and adipogenic differentiation/conversion. Interestingly, heparin did not only significantly increase mRNA expression and enzyme activity of the osteogenic marker alkaline phosphatase (ALP), but it also promoted mineralization during osteogenic differentiation and conversion. Furthermore, the mRNA expression of the osteogenic marker bone morphogenic protein 4 (BMP4) was enhanced. In addition, heparin administration partly prevented adipogenic differentiation and conversion demonstrated by reduced lipid droplet formation along with a decreased expression of adipogenic markers. Moreover, luciferase reporter assays, inhibitor experiments and gene expression analyses revealed that heparin had putative permissive effects on osteogenic signaling via the BMP pathway and reduced the mRNA expression of the Wnt pathway inhibitors dickkopf 1 (DKK1) and sclerostin (SOST). Taken together, our data show a rather supportive than inhibitory effect of heparin on osteogenic hBMSC differentiation and conversion in vitro. Further studies will have to investigate the net effects of heparin administration on bone formation versus bone resorption in vivo to unravel the molecular mechanisms of heparin-associated osteoporosis and reconcile conflicting experimental data with clinical observations.


Asunto(s)
Adipogénesis , Células de la Médula Ósea/citología , Heparina/química , Células Madre Mesenquimatosas/citología , Osteogénesis , Osteoporosis/fisiopatología , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/citología , Adulto , Anciano , Anticoagulantes/química , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Linaje de la Célula , Femenino , Marcadores Genéticos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lípidos/química , Masculino , Persona de Mediana Edad , Osteocitos/citología , Osteoporosis/etiología , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo
10.
J Steroid Biochem Mol Biol ; 139: 252-61, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23262262

RESUMEN

Phytoestrogens and other plant-derived compounds and extracts have been developed for the treatment of menopause-related complaints and disorders, e.g. hot flushes and osteoporosis. Since estrogens have been discussed to enhance the risk for hormone-sensitive cancers, research activities try to find alternatives. Phytoestrogens like genistein and resveratrol as well as other plant-derived compounds are capable of substituting for estrogens to some extent. Their effects on mesenchymal stem cells and the tissues derived therefrom have been investigated in vitro and in preclinical settings. Besides their well-known estrogenic, i.e. mainly antiresorptive effects on bone via estrogen receptor (ER) signalling, they also directly or indirectly affect osteogenic and adipogenic pathways. As a novel mechanism, phytoestrogens and plant-derived saponins and flavonoids like kaempferol and xanthohumol have been described to reciprocally affect the osteogenic versus the adipogenic differentiation pathway. Both, ER-mediated and other pathways mediate a shift towards osteogenesis by inhibiting PPARγ and C/EBPα, the key adipogenic transcription factors (TFs), while stimulating the key osteogenic TFs Runx2 and Sp7. Besides ER signalling, the broad spectrum of molecular mechanisms supporting osteogenesis comprises the modulation of PPARγ, Wnt/ß-catenin, and Sirt1 signalling, which inversely influence the transcription or transactivation of osteogenic versus adipogenic TFs. Preventing the age- and hormone deficiency-related shift towards adipogenesis without provoking adverse estrogenic effects represents a very promising strategy for treating bone loss and other metabolic diseases beyond bone. Research on plant-derived compounds will have to be pursued in vitro as well as in preclinical studies and controlled clinical trials in humans are urgently needed. This article is part of a Special Issue entitled 'Phytoestrogens'.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas/fisiología , Fitoestrógenos/farmacología , Adipogénesis/efectos de los fármacos , Animales , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Reguladores del Crecimiento de las Plantas/farmacología , Receptores de Estrógenos/metabolismo , Transducción de Señal
11.
Gene ; 551(2): 243-54, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25200494

RESUMEN

WNT-induced secreted protein 1 (WISP1/CCN4), a member of the CCN protein family, acts as a downstream factor of the canonical WNT signaling pathway. Its expression is known to affect proliferation and differentiation of human mesenchymal stromal cells (hMSCs), which are fundamental for the development and maintenance of the musculoskeletal system. Whereas a dysregulated, excessive expression of WISP1 often reflects its oncogenic potential via the inhibition of apoptosis, our study emphasizes the importance of WISP1 signaling for the survival of primary human cells. We have established the efficient and specific down-regulation of endogenous WISP1 transcripts by gene silencing in hMSCs and observed cell death as a consequence of WISP1 deficiency. This was confirmed by Annexin V staining for apoptotic cells. DNA microarray analyses of WISP1 down-regulated versus control samples revealed several clusters of differentially expressed genes important for apoptosis induction such as TNF-related apoptosis-inducing ligand 1 (TRAIL) and the corresponding apoptosis-inducing receptors TRAIL-R1 and -R2. An increased expression of TRAIL and its receptors TRAIL-R1 and -R2 in WISP1-deficient hMSCs was confirmed by immunocytofluorescence. Accordingly, WISP1 deficiency is likely to cause TRAIL-induced apoptosis. This is an important novel finding, which suggests that WISP1 is indispensable for the protection of healthy hMSCs against TRAIL-induced apoptosis.


Asunto(s)
Proteínas CCN de Señalización Intercelular/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas/genética , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas CCN de Señalización Intercelular/metabolismo , Supervivencia Celular/genética , Células Cultivadas , Ontología de Genes , Células HEK293 , Humanos , Peróxido de Hidrógeno/farmacología , Células Madre Mesenquimatosas/citología , Microscopía Confocal , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidantes/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
PLoS One ; 7(9): e45142, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028809

RESUMEN

Primary osteoporosis is an age-related disease characterized by an imbalance in bone homeostasis. While the resorptive aspect of the disease has been studied intensely, less is known about the anabolic part of the syndrome or presumptive deficiencies in bone regeneration. Multipotent mesenchymal stem cells (MSC) are the primary source of osteogenic regeneration. In the present study we aimed to unravel whether MSC biology is directly involved in the pathophysiology of the disease and therefore performed microarray analyses of hMSC of elderly patients (79-94 years old) suffering from osteoporosis (hMSC-OP). In comparison to age-matched controls we detected profound changes in the transcriptome in hMSC-OP, e.g. enhanced mRNA expression of known osteoporosis-associated genes (LRP5, RUNX2, COL1A1) and of genes involved in osteoclastogenesis (CSF1, PTH1R), but most notably of genes coding for inhibitors of WNT and BMP signaling, such as Sclerostin and MAB21L2. These candidate genes indicate intrinsic deficiencies in self-renewal and differentiation potential in osteoporotic stem cells. We also compared both hMSC-OP and non-osteoporotic hMSC-old of elderly donors to hMSC of ∼30 years younger donors and found that the transcriptional changes acquired between the sixth and the ninth decade of life differed widely between osteoporotic and non-osteoporotic stem cells. In addition, we compared the osteoporotic transcriptome to long term-cultivated, senescent hMSC and detected some signs for pre-senescence in hMSC-OP.Our results suggest that in primary osteoporosis the transcriptomes of hMSC populations show distinct signatures and little overlap with non-osteoporotic aging, although we detected some hints for senescence-associated changes. While there are remarkable inter-individual variations as expected for polygenetic diseases, we could identify many susceptibility genes for osteoporosis known from genetic studies. We also found new candidates, e.g. MAB21L2, a novel repressor of BMP-induced transcription. Such transcriptional changes may reflect epigenetic changes, which are part of a specific osteoporosis-associated aging process.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Osteoporosis/genética , Osteoporosis/patología , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Densidad Ósea/genética , Senescencia Celular/genética , Análisis por Conglomerados , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Fracturas Osteoporóticas/genética , Factores de Riesgo
13.
Exp Hematol ; 38(6): 504-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20347926

RESUMEN

OBJECTIVE: The identification of signaling pathways involved in megakaryocytopoiesis is essential for development of novel therapeutics to treat hematological disorders. Following our previous findings that megakaryocytes express functional channel-forming N-methyl-D-aspartate-type glutamate receptors, here we aimed to determine the glutamate release capacity in undifferentiated and differentiated megakaryocytes and the role of soluble N-ethyl maleimide-sensitive factor attachment protein receptor (SNARE) proteins that are known to be associated with vesicular exocytosis. MATERIALS AND METHODS: Using the megakaryocytic cell line MEG-01, primary megakaryocytes, and tissue sections of bone marrow, reverse transcription polymerase chain reaction, Western blot analysis, and immunolocalization were employed to detect factors required for vesicular glutamate release. Vesicle recycling was monitored by acridine orange and FM1-43 staining and glutamate release activity was assessed by an enzyme-linked fluorimetric assay. Genetically modified MEG-01 cells, with deletion or overexpression of SNARE and vesicular proteins, were also examined for glutamate release activity. RESULTS: We demonstrated that megakaryocytes express numerous proteins required for vesicular glutamate release, including core SNARE proteins, vesicle-associated membrane protein, soluble N-ethyl maleimide-sensitive factor attachment protein-23, and syntaxin, as well as specific glutamate-loading vesicle proteins, VGLUT1 and VGLUT2. Moreover, active vesicle recycling and differentiation-dependent glutamate release were observed in megakaryocytes. Vesicle-associated membrane protein-deficient MEG-01 cells, which are impaired in vesicle recycling, showed a 30% decrease in released glutamate, whereas overexpression of VGLUT1 exhibited up to a 2.2-fold increase in glutamate release. CONCLUSION: These data show that glutamate release from megakaryocytes occurs in a SNARE-dependent, exocytotic manner and is increased during differentiation, suggesting that manipulation of glutamate signaling could influence megakaryocytopoiesis and, therefore, offer a suitable target for the treatment of thrombosis and other hematological disorders.


Asunto(s)
Ácido Glutámico/metabolismo , Megacariocitos/metabolismo , Proteínas SNARE/fisiología , Animales , Secuencia de Bases , Western Blotting , Diferenciación Celular , Línea Celular , Cartilla de ADN , Exocitosis , Humanos , Megacariocitos/citología , Ratones
14.
Stem Cells Dev ; 19(5): 679-91, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19895235

RESUMEN

Pancreatic islet beta-cell replenishment can be driven by epithelial cells from exocrine pancreas via epithelial-mesenchymal transition (EMT) and the reverse process MET, while specified pancreatic mesenchymal cells control islet cell development and maintenance. The role of human islet-derived precursor cells (hIPCs) in regeneration and support of endocrine islets is under investigation. Here, we analyzed hIPCs as to their immunophenotype, multilineage differentiation capacity, and gene profiling, in comparison to human bone marrow-derived mesenchymal stem cells (hBM-MSCs). hIPCs and hBM-MSCs display a common mesenchymal character and express lineage-specific marker genes upon induction toward pancreatic endocrine and mesenchymal pathways of differentiation. hIPCs can go further along endocrine pathways while lacking some core mesenchymal differentiation attributes. Significance analysis of microarray (SAM) from 5 hBM-MSC and 3 hIPC donors mirrored such differences. Candidate gene cluster analysis disclosed differential expression of key lineage regulators, indicated a HoxA gene-associated positional memory in hIPCs and hBM-MSCs, and showed as well a clear transition state from mesenchyme to epithelium or vice versa in hIPCs. Our findings raise new research platforms to further clarify the potential of hIPCs to undergo complete MET thus contributing to islet cell replenishment, maintenance, and function.


Asunto(s)
Células de la Médula Ósea/citología , Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Regeneración , Células Madre/citología , Células Epiteliales/citología , Humanos , Páncreas Exocrino/citología , Células Madre/fisiología
15.
Expert Rev Endocrinol Metab ; 2(1): 59-78, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30743749

RESUMEN

Bone development, regeneration and maintenance are governed by osteogenic differentiation processes from mesenchymal stem cells through to mature bone cells, which are directed by local growth and differentiation factors and modulated strongly by hormones. Mesenchymal stem cells develop from both mesoderm and neural crest and can give rise to development, regeneration and maintenance of mesenchymal tissues, such as bone, cartilage, muscle, tendons and discs. There are only limited data regarding the effects of hormones on early events, such as regulation of stemness and maintenance of the mesenchymal stem cell pool. Hormones, such as estrogens, vitamin D-hormone and parathyroid hormone, besides others, are important modulators of osteogenic differentiation processes and bone formation, starting off with fate decision and the development of osteogenic offspring from mesenchymal stem cells, which end up in osteoblasts and osteocytes. Hormones are involved in fetal bone development and regeneration and, in childhood, adolescence and adulthood, they control adaptive needs for growth and reproduction, nutrition, physical power and crisis adaptation. As in other tissues, aging in mesenchymal stem cells and their osteogenic offspring is accompanied by the accumulation of genomic and proteomic damage caused by oxidative burden and insufficient repair. Failsafe programs, such as apoptosis and cellular senescence avoid tumorigenesis. Hormones can influence the pace of such events, thus supporting the quality of tissue regeneration in aging organisms in vivo; for example, by delaying osteoporosis development. The potential for hormones in systemic therapeutic strategies is well appreciated and some concepts are approved for clinical use already. Their potential for cell-based therapeutic strategies for tissue regeneration is probably underestimated and could enhance the quality of tissue-engineering constructs for transplantation and the concept of in situ-guided tissue regeneration.

16.
Biochem Biophys Res Commun ; 363(1): 209-15, 2007 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-17868648

RESUMEN

High glucose (HG) concentrations impair cellular functions and induce apoptosis. Exposition of mesenchymal stem cells (MSC) to HG was reported to reduce colony forming activity and induce premature senescence. We characterized the effects of HG on human MSC in vitro using telomerase-immortalized MSC (hMSC-TERT) and primary MSC (hMSC). HG (25mM) enhanced hMSC-TERT proliferation in long-term studies in contrast to hMSC where proliferation was unchanged. Thioredoxin-interacting protein, which is involved in apoptosis regulation, was stimulated by glucose in hMSC-TERT. However, apoptosis was not influenced by HG in both cell types. MSC treatment with HG favored osteogenic differentiation. MSC are resistant to HG toxicity, depending on the stemness of MSC. Proliferation and osteogenic differentiation are stimulated by HG. Effects of HG on the transient amplifying compartment of MSC may differ from those in mature cells. Further research is needed to unravel the molecular mechanisms of HG resistance of MSC.


Asunto(s)
Proteínas Portadoras/metabolismo , Glucosa/administración & dosificación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA