Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 320: 113011, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31306642

RESUMEN

Aberrant expression of DUX4, a gene unique to humans and primates, causes Facioscapulohumeral Muscular Dystrophy-1 (FSHD), yet the pathogenic mechanism is unknown. As transgenic overexpression models have largely failed to replicate the genetic changes seen in FSHD, many studies of endogenously expressed DUX4 have been limited to patient biopsies and myogenic cell cultures, which never fully differentiate into mature muscle fibers. We have developed a method to xenograft immortalized human muscle precursor cells from patients with FSHD and first-degree relative controls into the tibialis anterior muscle compartment of immunodeficient mice, generating human muscle xenografts. We report that FSHD cells mature into organized and innervated human muscle fibers with minimal contamination of murine myonuclei. They also reconstitute the satellite cell niche within the xenografts. FSHD xenografts express DUX4 and DUX4 downstream targets, retain the 4q35 epigenetic signature of their original donors, and express a novel protein biomarker of FSHD, SLC34A2. Ours is the first scalable, mature in vivo human model of FSHD. It should be useful for studies of the pathogenic mechanism of the disease as well as for testing therapeutic strategies targeting DUX4 expression.


Asunto(s)
Modelos Animales de Enfermedad , Xenoinjertos , Distrofia Muscular Facioescapulohumeral , Mioblastos/trasplante , Animales , Proteínas de Homeodominio/genética , Humanos , Ratones , Músculo Esquelético/patología , Distrofia Muscular Facioescapulohumeral/genética
2.
Nat Commun ; 9(1): 3583, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30181605

RESUMEN

Enhanced telomere maintenance is evident in malignant cancers. While telomeres are thought to be inherently heterochromatic, detailed mechanisms of how epigenetic modifications impact telomere protection and structures are largely unknown in human cancers. Here we develop a molecular tethering approach to experimentally enrich heterochromatin protein HP1α specifically at telomeres. This results in increased deposition of H3K9me3 at cancer cell telomeres. Telomere extension by telomerase is attenuated, and damage-induced foci at telomeres are reduced, indicating augmentation of telomere stability. Super-resolution STORM imaging shows an unexpected increase in irregularity of telomeric structure. Telomere-tethered chromo shadow domain (CSD) mutant I165A of HP1α abrogates both the inhibition of telomere extension and the irregularity of telomeric structure, suggesting the involvement of at least one HP1α-ligand in mediating these effects. This work presents an approach to specifically manipulate the epigenetic status locally at telomeres to uncover insights into molecular mechanisms underlying telomere structural dynamics.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Telómero/metabolismo , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Daño del ADN , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Microscopía/métodos , Mutación , Dominios Proteicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Telómero/genética , Telómero/ultraestructura , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
3.
PLoS Biol ; 14(12): e2000016, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977688

RESUMEN

Telomerase is expressed in early human development and then becomes silenced in most normal tissues. Because ~90% of primary human tumors express telomerase and generally maintain very short telomeres, telomerase is carefully regulated, particularly in large, long-lived mammals. In the current report, we provide substantial evidence for a new regulatory control mechanism of the rate limiting catalytic protein component of telomerase (hTERT) that is determined by the length of telomeres. We document that normal, young human cells with long telomeres have a repressed hTERT epigenetic status (chromatin and DNA methylation), but the epigenetic status is altered when telomeres become short. The change in epigenetic status correlates with altered expression of TERT and genes near to TERT, indicating a change in chromatin. Furthermore, we identified a chromosome 5p telomere loop to a region near TERT in human cells with long telomeres that is disengaged with increased cell divisions as telomeres progressively shorten. Finally, we provide support for a role of the TRF2 protein, and possibly TERRA, in the telomere looping maintenance mechanism through interactions with interstitial TTAGGG repeats. This provides new insights into how the changes in genome structure during replicative aging result in an increased susceptibility to age-related diseases and cancer prior to the initiation of a DNA damage signal.


Asunto(s)
Envejecimiento/genética , Regulación Enzimológica de la Expresión Génica , Neoplasias/genética , Telomerasa/genética , Telómero , Animales , Cromosomas Humanos Par 5 , Humanos , Primates/genética
4.
Skelet Muscle ; 6: 4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925213

RESUMEN

BACKGROUND: Studies of the pathogenic mechanisms underlying human myopathies and muscular dystrophies often require animal models, but models of some human diseases are not yet available. Methods to promote the engraftment and development of myogenic cells from individuals with such diseases in mice would accelerate such studies and also provide a useful tool for testing therapeutics. Here, we investigate the ability of immortalized human myogenic precursor cells (hMPCs) to form mature human myofibers following implantation into the hindlimbs of non-obese diabetic-Rag1 (null) IL2rγ (null) (NOD-Rag)-immunodeficient mice. RESULTS: We report that hindlimbs of NOD-Rag mice that are X-irradiated, treated with cardiotoxin, and then injected with immortalized control hMPCs or hMPCs from an individual with facioscapulohumeral muscular dystrophy (FSHD) develop mature human myofibers. Furthermore, intermittent neuromuscular electrical stimulation (iNMES) of the peroneal nerve of the engrafted limb enhances the development of mature fibers in the grafts formed by both immortal cell lines. With control cells, iNMES increases the number and size of the human myofibers that form and promotes closer fiber-to-fiber packing. The human myofibers in the graft are innervated, fully differentiated, and minimally contaminated with murine myonuclei. CONCLUSIONS: Our results indicate that control and FSHD human myofibers can form in mice engrafted with hMPCs and that iNMES enhances engraftment and subsequent development of mature human muscle.


Asunto(s)
Diferenciación Celular , Estimulación Eléctrica/métodos , Desarrollo de Músculos , Distrofia Muscular Facioescapulohumeral/patología , Mioblastos Esqueléticos/trasplante , Unión Neuromuscular , Nervio Peroneo , Adulto , Animales , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Supervivencia de Injerto , Xenoinjertos , Humanos , Masculino , Ratones Endogámicos NOD , Ratones Transgénicos , Distrofia Muscular Facioescapulohumeral/metabolismo , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patología , Factores de Tiempo
5.
Genome Res ; 25(12): 1781-90, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26359233

RESUMEN

DNA is organized into complex three-dimensional chromatin structures, but how this spatial organization regulates gene expression remains a central question. These DNA/chromatin looping structures can range in size from 10-20 kb (enhancers/repressors) to many megabases during intra- and inter-chromosomal interactions. Recently, the influence of telomere length on chromatin organization prior to senescence has revealed the existence of long-distance chromatin loops that dictate the expression of genes located up to 10 Mb from the telomeres (Telomere Position Effect-Over Long Distances [TPE-OLD]). Here, we demonstrate the existence of a telomere loop at the 4q35 locus involving the sorbin and SH3 domain-containing protein 2 gene, SORBS2, a skeletal muscle protein using a modification of the chromosome conformation capture method. The loop reveals a cis-acting mechanism modifying SORBS2 transcription. The expression of this gene is altered by TPE-OLD in myoblasts from patients affected with the age-associated genetic disease, facioscapulohumeral muscular dystrophy (FSHD1A, MIM 158900). SORBS2 is expressed in FSHD myoblasts with short telomeres, while not detectable in FSHD myoblasts with long telomeres or in healthy myoblasts regardless of telomere length. This indicates that TPE-OLD may modify the regulation of the 4q35 locus in a pathogenic context. Upon differentiation, both FSHD and healthy myotubes express SORBS2, suggesting that SORBS2 is normally up-regulated by maturation/differentiation of skeletal muscle and is misregulated by TPE-OLD-dependent variegation in FSHD myoblasts. These findings provide additional insights for the complexity and age-related symptoms of FSHD.


Asunto(s)
Proteínas de Homeodominio/genética , Células Musculares/metabolismo , Distrofia Muscular Facioescapulohumeral/genética , Acortamiento del Telómero , Telómero/genética , Activación Transcripcional , Proteínas Adaptadoras Transductoras de Señales , Biopsia , Deleción Cromosómica , Cromosomas Humanos Par 4 , Metilación de ADN , Epistasis Genética , Regulación de la Expresión Génica , Sitios Genéticos , Proteínas de Homeodominio/metabolismo , Humanos , Hibridación Fluorescente in Situ , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Facioescapulohumeral/metabolismo , Distrofia Muscular Facioescapulohumeral/patología , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos , Proteínas de Unión al ARN
6.
Genes Dev ; 28(22): 2464-76, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25403178

RESUMEN

While global chromatin conformation studies are emerging, very little is known about the chromatin conformation of human telomeres. Most studies have focused on the role of telomeres as a tumor suppressor mechanism. Here we describe how telomere length regulates gene expression long before telomeres become short enough to produce a DNA damage response (senescence). We directly mapped the interactions adjacent to specific telomere ends using a Hi-C (chromosome capture followed by high-throughput sequencing) technique modified to enrich for specific genomic regions. We demonstrate that chromosome looping brings the telomere close to genes up to 10 Mb away from the telomere when telomeres are long and that the same loci become separated when telomeres are short. Furthermore, expression array analysis reveals that many loci, including noncoding RNAs, may be regulated by telomere length. We report three genes (ISG15 [interferon-stimulated gene 15 kd], DSP [Desmoplakin], and C1S [complement component 1s subcomplement]) located at three different subtelomeric ends (1p, 6p, and 12p) whose expressions are altered with telomere length. Additionally, we confirmed by in situ analysis (3D-FISH [three-dimensional fluorescence in situ hybridization]) that chromosomal looping occurs between the loci of those genes and their respective telomere ends. We term this process TPE-OLD for "telomere position effect over long distances." Our results suggest a potential novel mechanism for how telomere shortening could contribute to aging and disease initiation/progression in human cells long before the induction of a critical DNA damage response.


Asunto(s)
Regulación de la Expresión Génica , Acortamiento del Telómero/genética , Telómero/genética , Telómero/metabolismo , Células Cultivadas , Cromatina/metabolismo , Perfilación de la Expresión Génica , Humanos , Mioblastos/citología
7.
Cell Tissue Bank ; 15(2): 213-25, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24828570

RESUMEN

Human amniotic membrane (hAM) represents a tissue that is well established as biomaterial in the clinics with potential for new applications in regenerative medicine. For tissue engineering (TE) strategies, cells are usually combined with inductive factors and a carrier substrate. We have previously recognized that hAM represents a natural, preformed sheet including highly potent stem cells. In the present approach for cartilage regeneration we have induced chondrogenesis in hAM in vitro. For this, hAM biopsies were cultured for up to 56 days under chondrogenic conditions. The induced hAM was characterized for remaining viability, glycosaminoglycan (GAG) accumulation using histochemical analysis, and a quantitative assay. Collagen I, II and X was immunohistochemically determined and cartilage-specific mRNA expression of (sex determining region Y-) box 9, cartilage oligomeric matrix protein (COMP), aggrecan (AGC1), versican (CSPG2), COL1A1, COL9A2, melanoma inhibitory activity (MIA), and cartilage-linking protein 1 (CRTL1) analyzed by quantitative real-time polymerase chain reaction. Human AM was successfully induced to accumulate GAG, as demonstrated by Alcianblue staining and a significant (p < 0.001) increase of GAG/viability under chondrogenic conditions peaking in a 29.9 ± 0.9-fold induction on day 56. Further, upon chondrogenic induction collagen II positive areas were identified within histological sections and cartilage-specific markers including COMP, AGC1, CSPG2, COL1A1, COL9A2, MIA, and CRTL1 were found upregulated at mRNA level. This is the first study, demonstrating that upon in vitro induction viable human amnion expresses cartilage-specific markers and accumulates GAGs within the biomatrix. This is a promising first step towards a potential use of living hAM for cartilage TE.


Asunto(s)
Amnios/citología , Diferenciación Celular , Linaje de la Célula/fisiología , Condrogénesis/fisiología , Placenta/citología , Cartílago/citología , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Embarazo , Células Madre/citología
8.
Skelet Muscle ; 3(1): 28, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24314268

RESUMEN

BACKGROUND: Congenital muscular dystrophy Type 1A (MDC1A) is a severe, recessive disease of childhood onset that is caused by mutations in the LAMA2 gene encoding laminin-α2. Studies with both mouse models and primary cultures of human MDC1A myogenic cells suggest that aberrant activation of cell death is a significant contributor to pathogenesis in laminin-α2-deficiency. METHODS: To overcome the limited population doublings of primary cultures, we generated immortalized, clonal lines of human MDC1A myogenic cells via overexpression of both CDK4 and the telomerase catalytic component (human telomerase reverse transcriptase (hTERT)). RESULTS: The immortalized MDC1A myogenic cells proliferated indefinitely when cultured at low density in high serum growth medium, but retained the capacity to form multinucleate myotubes and express muscle-specific proteins when switched to low serum medium. When cultured in the absence of laminin, myotubes formed from immortalized MDC1A myoblasts, but not those formed from immortalized healthy or disease control human myoblasts, showed significantly increased activation of caspase-3. This pattern of aberrant caspase-3 activation in the immortalized cultures was similar to that found previously in primary MDC1A cultures and laminin-α2-deficient mice. CONCLUSIONS: Immortalized MDC1A myogenic cells provide a new resource for studies of pathogenetic mechanisms and for screening possible therapeutic approaches in laminin-α2-deficiency.

9.
Nat Struct Mol Biol ; 20(6): 671-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23644600

RESUMEN

Telomeres may regulate human disease by at least two independent mechanisms. First, replicative senescence occurs once short telomeres generate DNA-damage signals that produce a barrier to tumor progression. Second, telomere position effects (TPE) could change gene expression at intermediate telomere lengths in cultured human cells. Here we report that telomere length may contribute to the pathogenesis of facioscapulohumeral muscular dystrophy (FSHD). FSHD is a late-onset disease genetically residing only 25-60 kilobases from the end of chromosome 4q. We used a floxable telomerase to generate isogenic clones with different telomere lengths from affected patients and their unaffected siblings. DUX4, the primary candidate for FSHD pathogenesis, is upregulated over ten-fold in FSHD myoblasts and myotubes with short telomeres, and its expression is inversely proportional to telomere length. FSHD may be the first known human disease in which TPE contributes to age-related phenotype.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Distrofia Muscular Facioescapulohumeral/genética , Telómero/metabolismo , Células Cultivadas , Humanos , Distrofia Muscular Facioescapulohumeral/patología , Mioblastos/fisiología , Regulación hacia Arriba
10.
Rare Dis ; 1: e26142, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25003004

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is a progressive myopathy with a relatively late age of onset (usually in the late teens) compared with Duchenne and many other muscular dystrophies. The current FSHD disease model postulates that contraction of the D4Z4 array at chromosome 4q35 leads to a more open chromatin conformation in that region and allows transcription of the DUX4 gene. DUX4 mRNA is stable only when transcribed from certain haplotypes that contain a polyadenylation signal. DUX4 protein is hypothesized to cause FSHD by mediating cytotoxicity and impairing skeletal muscle differentiation. We recently showed in a cell culture model that DUX4 expression is regulated by telomere length, suggesting that telomere shortening during aging may be partially responsible for the delayed onset and progressive nature of FSHD. We here put our data in the context of other recent findings arguing that progressive telomere shortening may play a critical role in FSHD but is not the whole story and that the current disease model needs additional refinement.

11.
J Orthop Res ; 30(3): 489-96, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22371968

RESUMEN

Mineralized scaffolds are widely used as bone grafts with the assumption that bone marrow derived cells colonize and remodel them. This process is slow and often unreliable so we aimed to improve the biocompatibility of bone grafts by pre-seeding them with human mesenchymal stem cells from either bone marrow or dental pulp. Under standard cell culture conditions very low number of seeded cells remained on the surface of freeze-dried human or bovine bone graft or hydroxyapatite. Coating the scaffolds with fibronectin or collagen improved seeding efficiency but the cells failed to grow on the surface until the 18th day. In contrast, human albumin was a very potent facilitator of both seeding and proliferation on allografts which was further improved by culturing in a rotating bioreactor. Electron microscopy revealed that cells do not form a monolayer but span the pores, emphasizing the importance of pore size and microstructure. Albumin coated bone chips were able to unite a rat femoral segmental defect, while uncoated ones did not. Micro-hardness measurements confirmed that albumin coating does not influence the physical characteristics of the scaffold, so it is possible to introduce albumin coating into the manufacturing process of lyophilized bone allografts.


Asunto(s)
Trasplante Óseo , Colágeno Tipo I/fisiología , Fibronectinas/fisiología , Células Madre Mesenquimatosas/fisiología , Albúmina Sérica/fisiología , Adolescente , Adulto , Animales , Reactores Biológicos , Células de la Médula Ósea/fisiología , Adhesión Celular , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Pulpa Dental/citología , Liofilización , Humanos , Masculino , Ratas , Ratas Wistar , Porcinos , Andamios del Tejido , Trasplante Homólogo , Adulto Joven
12.
J Biol Chem ; 286(50): 43294-300, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22020932

RESUMEN

Normal human colonic epithelial cells (HCECs) are not immortalized by telomerase alone but also require CDK4. Some human cell types growth-arrest due to stress- or aberrant signaling-induced senescence (stasis). Stasis represents the consequences of growth conditions culture that are inadequate to maintain long-term proliferation. Overexpressed CDK4 titers out p16 and allows cells to ignore the growth arrest signals produced by stasis. To identify factors contributing to the inadequate culture environment, we used a 62,000-member shRNA library to knock down factors cooperating with human telomerase reverse transcriptase (hTERT) in the immortalization of HCECs. Knockdown of Klotho gamma (KLG; also known as KLPH and LCTL) allowed hTERT to immortalize HCECs. KLG is one isoform of the Klotho family of factors that coordinate interaction between different FGF ligands and the FGF receptor. We also found that knockdown of KLG induced another member of the Klotho family, Klotho beta (KLB). Induction of KLB was maintained and could activate ERK1/2 in immortalized cells. Supplementation of the culture medium with the KLB ligand FGF19 had a similar effect on hTERT-expressing HCECs as knockdown of KLG regarding both immortalization and down-regulation of the tumor suppressor Klotho alpha. Together, these data suggest that KLB is an important regulator in the immortalization of HCECs by facilitating FGF19 growth factor signaling.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Colon/citología , Células Epiteliales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , ARN Interferente Pequeño/fisiología , Western Blotting , Línea Celular , Factores de Crecimiento de Fibroblastos/farmacología , Glucuronidasa/genética , Humanos , Proteínas Klotho , ARN Interferente Pequeño/genética , Telomerasa/genética , Telomerasa/metabolismo
13.
Skelet Muscle ; 1(1): 12, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21798090

RESUMEN

BACKGROUND: A hallmark of muscular dystrophies is the replacement of muscle by connective tissue. Muscle biopsies from patients severely affected with facioscapulohumeral muscular dystrophy (FSHD) may contain few myogenic cells. Because the chromosomal contraction at 4q35 linked to FSHD is thought to cause a defect within myogenic cells, it is important to study this particular cell type, rather than the fibroblasts and adipocytes of the endomysial fibrosis, to understand the mechanism leading to myopathy. RESULTS: We present a protocol to establish clonal myogenic cell lines from even severely dystrophic muscle that has been replaced mostly by fat, using overexpression of CDK4 and the catalytic component of telomerase (human telomerase reverse transcriptase; hTERT), and a subsequent cloning step. hTERT is necessary to compensate for telomere loss during in vitro cultivation, while CDK4 prevents a telomere-independent growth arrest affecting CD56+ myogenic cells, but not their CD56- counterpart, in vitro. CONCLUSIONS: These immortal cell lines are valuable tools to reproducibly study the effect of the FSHD mutation within myoblasts isolated from muscles that have been severely affected by the disease, without the confounding influence of variable amounts of contaminating connective-tissue cells.

14.
Mol Cell ; 42(3): 297-307, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21549308

RESUMEN

Specific information about how telomerase acts in vivo is necessary for understanding telomere dynamics in human tumor cells. Our results imply that, under homeostatic telomere length-maintenance conditions, only one molecule of telomerase acts at each telomere during every cell division and processively adds ∼60 nt to each end. In contrast, multiple molecules of telomerase act at each telomere when telomeres are elongating (nonequilibrium conditions). Telomerase extension is less processive during the first few weeks following the reversal of long-term treatment with the telomerase inhibitor Imetelstat (GRN163L), a time when Cajal bodies fail to deliver telomerase RNA to telomeres. This result implies that processing of telomerase by Cajal bodies may affect its processivity. Overexpressed telomerase is also less processive than the endogenously expressed telomerase. These findings reveal two major distinct extension modes adopted by telomerase in vivo.


Asunto(s)
Homeostasis , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , Western Blotting , Línea Celular Tumoral , Cuerpos Enrollados/metabolismo , Fase G1 , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Modelos Genéticos , Oligonucleótidos/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fase S , Telomerasa/antagonistas & inhibidores , Telomerasa/genética
15.
Biomaterials ; 31(33): 8659-65, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20719379

RESUMEN

Tissue engineering strategies usually require cell isolation and combination with a suitable biomaterial. Human amniotic membrane (AM) represents a natural two-layered sheet comprising cells with proven stem cell characteristics. In our approach, we evaluated the differentiation potential of AM in toto with its sessile stem cells as alternative to conventional approaches requiring cell isolation and combination with biomaterials. For this, AM-biopsies were differentiated in vitro using two osteogenic media compared with control medium (CM) for 28 days. Mineralization and osteocalcin expression was demonstrated by (immuno)histochemistry. Alkaline phosphatase (AP) activity, calcium contents and mRNA expression of RUNX2, AP, osteopontin, osteocalcin, BMP-2 (bone morphogenetic protein), and BMP-4 were quantified and AM viability was evaluated. Under osteogenic conditions, AM-biopsies mineralized successfully and by day 28 the majority of cells expressed osteocalcin. This was confirmed by a significant rise in calcium contents (up to 27.4 ± 6.8 mg/dl d28), increased AP activity, and induction of RUNX2, AP, BMP-2 and BMP-4 mRNA expression. Relatively high levels of viability were retained, especially in osteogenic media (up to 78.3 ± 19.0% d14; 62.9 ± 22.3% d28) compared to CM (42.2 ± 15.2% d14; 35.1 ± 8.6% d28). By this strategy, stem cells within human AM can successfully be driven along the osteogenic pathways while residing within their natural environment.


Asunto(s)
Amnios/citología , Diferenciación Celular , Osteogénesis , Fosfatasa Alcalina/metabolismo , Amnios/efectos de los fármacos , Amnios/enzimología , Biomarcadores/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/enzimología , Osteogénesis/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Tissue Eng Part A ; 15(7): 1843-54, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19125642

RESUMEN

Cell banking of mesenchymal stem cells (SCs) from various human tissues has significantly increased the feasibility of SC-based therapies. Sources such as adipose tissue and amnion offer outstanding possibilities for allogeneic transplantation due to their high differentiation potential and their ability to modulate immune reaction. Limitations, however, concern the reduced replicative potential as a result of progressive telomere erosion, which hampers scaleable production and long-term analysis of these cells. Here we report the establishment and characterization of two human amnion-derived and two human adipose-derived SC lines immortalized by ectopic expression of the catalytic subunit of human telomerase (hTERT). hTERT overexpression resulted in continuously growing SC lines that were largely unaltered concerning surface marker profile, morphology, karyotype, and immunosuppressive capacity with similar or enhanced differentiation potential for up to 87 population doublings. While all generated lines showed equal immunomodulation compared to the parental cells, one of the amnion-derived immortalized lines resulted in significantly increased immunogenicity. Although telomerase proves as important tool for immortalizing cells, our data emphasize the need for careful and standardized characterization of each individual cell population for cell banks.


Asunto(s)
Tejido Adiposo/citología , Amnios/citología , Diferenciación Celular , Factores Inmunológicos/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Telomerasa/metabolismo , Adipogénesis , Fosfatasa Alcalina/metabolismo , Antígenos de Superficie/metabolismo , Recuento de Células , Línea Celular Transformada , Proliferación Celular , Forma de la Célula , Humanos , Cariotipificación , Leucocitos Mononucleares/inmunología , Células Madre Mesenquimatosas/enzimología , Neoplasias/patología , Osteocalcina/metabolismo , Osteogénesis , PPAR gamma/metabolismo , Transducción Genética
17.
Am J Physiol Renal Physiol ; 295(5): F1365-75, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18715936

RESUMEN

Telomere-dependent replicative senescence is one of the mechanisms that limit the number of population doublings of normal human cells. By overexpression of telomerase, cells of various origins have been successfully immortalized without changing the phenotype. While a limited number of telomerase-immortalized cells of epithelial origin are available, none of renal origin has been reported so far. Here we have established simple and safe conditions that allow serial passaging of renal proximal tubule epithelial cells (RPTECs) until entry into telomere-dependent replicative senescence. As reported for other cells, senescence of RPTECs is characterized by arrest in G1 phase, shortened telomeres, staining for senescence-associated beta-galactosidase, and accumulation of gamma-H2AX foci. Furthermore, ectopic expression of the catalytic subunit of telomerase (TERT) was sufficient to immortalize these cells. Characterization of immortalized RPTEC/TERT1 cells shows characteristic morphological and functional properties like formation of tight junctions and domes, expression of aminopeptidase N, cAMP induction by parathyroid hormone, sodium-dependent phosphate uptake, and the megalin/cubilin transport system. No genomic instability within up to 90 population doublings has been observed. Therefore, these cells are proposed as a valuable model system not only for cell biology but also for toxicology, drug screening, biogerontology, as well as tissue engineering approaches.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Telomerasa/genética , Antígenos CD13/metabolismo , Cadherinas/metabolismo , Ciclo Celular/fisiología , Línea Celular Transformada , Senescencia Celular/fisiología , Cilios/ultraestructura , AMP Cíclico/metabolismo , Células Epiteliales/citología , Células Epiteliales/fisiología , Histonas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Túbulos Renales Proximales/citología , Proteínas de la Membrana/metabolismo , Microscopía Electrónica , Microvellosidades/ultraestructura , Ocludina , Hormona Paratiroidea/farmacología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/metabolismo , Telomerasa/metabolismo , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Transfección , beta-Galactosidasa/metabolismo , gamma-Glutamiltransferasa/metabolismo
18.
Eur J Cancer ; 44(6): 866-75, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18296042

RESUMEN

Maintenance of telomere length has been reported to be an absolute requirement for unlimited growth of human tumour cells and in about 85% of cases, this is achieved by reactivation of telomerase, the enzyme that elongates telomeres. Only in rare cases, like in human medullary thyroid carcinomas (MTC), telomerase activity (TA) is low or undetectable; however, this does not limit tumours to become clinically significant. Here, we report that very low TA (below 5% of HEK293) observed in MTC cell strains derived from different patients, although not sufficient for immortalising the cells, is necessary for prolonging their replicative life span. Telomere erosion led to induction of a crisis period after long-term in vitro cultivation, which was reached earlier when treating the cells with MST-312, a telomerase inhibitor at non-toxic concentrations. Crisis was bypassed either by ectopic hTERT introduction or by infrequent spontaneous immortalisation, the latter of which was always associated with telomerase reactivation and changes of the cellular phenotype. While confirming the high importance of telomerase for tumour development, these data draw attention to the relevance of low TA: although insufficient for telomere stabilisation, it allows MTC cells to reach more population doublings, increasing both cell numbers as well as the risk of accumulating mutations and thus might support the development of clinically significant MTC.


Asunto(s)
Carcinoma Medular/enzimología , Proteínas de Neoplasias/metabolismo , Telomerasa/metabolismo , Neoplasias de la Tiroides/enzimología , Carcinoma Medular/patología , Progresión de la Enfermedad , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Telómero/patología , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas
19.
Stem Cells ; 26(2): 300-11, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17975221

RESUMEN

Placental tissue draws great interest as a source of cells for regenerative medicine because of the phenotypic plasticity of many of the cell types isolated from this tissue. Furthermore, placenta, which is involved in maintaining fetal tolerance, contains cells that display immunomodulatory properties. These two features could prove useful for future cell therapy-based clinical applications. Placental tissue is readily available and easily procured without invasive procedures, and its use does not elicit ethical debate. Numerous reports describing stem cells from different parts of the placenta, using nearly as numerous isolation and characterization procedures, have been published. Considering the complexity of the placenta, an urgent need exists to define, as clearly as possible, the region of origin and methods of isolation of cells derived from this tissue. On March 23-24, 2007, the first international Workshop on Placenta Derived Stem Cells was held in Brescia, Italy. Most of the research published in this area focuses on mesenchymal stromal cells isolated from various parts of the placenta or epithelial cells isolated from amniotic membrane. The aim of this review is to summarize and provide the state of the art of research in this field, addressing aspects such as cell isolation protocols and characteristics of these cells, as well as providing preliminary indications of the possibilities for use of these cells in future clinical applications.


Asunto(s)
Separación Celular/métodos , Células Madre Embrionarias/citología , Placenta/citología , Amnios/citología , Amnios/inmunología , Animales , Antígenos de Superficie/metabolismo , Adhesión Celular , Diferenciación Celular , Corion/citología , Corion/inmunología , Ensayo de Unidades Formadoras de Colonias , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Células Epiteliales/citología , Células Epiteliales/inmunología , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Tolerancia Inmunológica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ratones , Placenta/inmunología , Embarazo , Trasplante de Células Madre , Células del Estroma/citología , Células del Estroma/inmunología , Bancos de Tejidos , Trofoblastos/citología , Trofoblastos/inmunología
20.
Tissue Eng ; 13(6): 1173-83, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17518752

RESUMEN

Bone marrow-derived mesenchymal stem cells (BMSCs) have been used for allogeneic application in tissue engineering but have certain drawbacks. Therefore, stem cells (SC)s derived from other adult tissue sources have been considered as an alternative. However, there is only limited knowledge on their immunomodulatory properties. The aim of our study was to compare the immunomodulatory potential of human amniotic mesenchymal and human amniotic epithelial cells with that of human adipose-derived SCs under identical experimental conditions. We have demonstrated a dose-dependent inhibition of peripheral blood mononuclear cell (PBMC) immune responses in mixed lymphocyte reactions (up to 66-93% inhibition) and phytohemagglutinin activation assays (up to 67-96% inhibition). The lowest SC-to-PBMC ratio able to inhibit PBMC proliferation significantly was 1:8. Subcultivation (passage 2-6) did not alter immunoinhibitory properties, whereas cryopreservation significantly reduced the immunomodulatory potential. Using transwell systems, we have demonstrated an inhibition mechanism that is dependent on cell contact. Additionally, in coculture with allogeneic PBMCs, SCs were well tolerated and at most provoked mild alloreactions in singular cases. This study demonstrates, for the first time, contact- and dose-dependent immunosuppression of mesenchymal and epithelial amniotic SC populations, as well as of adipose tissue-derived SCs. All three cell types may be considered as possible alternatives to BMSCs for allogeneic application in tissue engineering.


Asunto(s)
Adipocitos/citología , Adipocitos/inmunología , Amnios/citología , Amnios/inmunología , Factores Inmunológicos/inmunología , Células Madre/citología , Células Madre/inmunología , Comunicación Celular/inmunología , Diferenciación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ingeniería de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...