Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
2.
N Engl J Med ; 380(9): 842-849, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30811910

RESUMEN

Sesquizygotic multiple pregnancy is an exceptional intermediate between monozygotic and dizygotic twinning. We report a monochorionic twin pregnancy with fetal sex discordance. Genotyping of amniotic fluid from each sac showed that the twins were maternally identical but chimerically shared 78% of their paternal genome, which makes them genetically in between monozygotic and dizygotic; they are sesquizygotic. We observed no evidence of sesquizygosis in 968 dizygotic twin pairs whom we screened by means of pangenome single-nucleotide polymorphism genotyping. Data from published repositories also show that sesquizygosis is a rare event. Detailed genotyping implicates chimerism arising at the juncture of zygotic division, termed heterogonesis, as the likely initial step in the causation of sesquizygosis.


Asunto(s)
Quimera , Fertilización , Gemelos Monocigóticos/genética , Adulto , Alelos , Embolia Paradójica/complicaciones , Femenino , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Embarazo , Embarazo Gemelar , Tromboembolia/etiología , Ultrasonografía Prenatal , Vena Cava Inferior
3.
Nanoscale ; 10(17): 8226-8239, 2018 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-29682654

RESUMEN

There is currently intense interest in new methods for understanding the fate of therapeutically-relevant cells, such as mesenchymal stem cells (MSCs). The absence of a confounding background signal and consequent unequivocal assignment makes 19F MRI one of the most attractive modalities for the tracking of injected cells in vivo. We describe here the synthesis of novel partly-fluorinated polymeric nanoparticles with small size and high fluorine content as MRI agents. The polymers, constructed from perfluoropolyether methacrylate (PFPEMA) and oligo(ethylene glycol) methacrylate (OEGMA) have favourable cell uptake profiles and excellent MRI performance. To facilitate cell studies the polymer was further conjugated with a fluorescent dye creating a dual-modal imaging agent. The efficacy of labelling of MSCs was assessed using 19F NMR, flow cytometry and confocal microscopy. The labelling efficiency of 2.6 ± 0.1 × 1012 19F atoms per cell, and viability of >90% demonstrates high uptake and good tolerance by the cells. This loading translates to a minimum 19F MRI detection sensitivity of ∼7.4 × 103 cells per voxel. Importantly, preliminary in vivo data demonstrate that labelled cells can be readily detected within a short acquisition scan period (12 minutes). Hence, these copolymers show outstanding potential for 19F MRI cellular tracking and for quantification of non-phagocytic and therapeutically-relevant cells in vivo.


Asunto(s)
Rastreo Celular , Medios de Contraste/química , Imagen por Resonancia Magnética con Fluor-19 , Células Madre Mesenquimatosas/citología , Animales , Femenino , Citometría de Flujo , Colorantes Fluorescentes , Flúor , Humanos , Ratones Endogámicos NOD , Ratones SCID , Microscopía Confocal
4.
Stem Cell Reports ; 10(3): 890-904, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29478891

RESUMEN

The existence of bipotential precursors for both mesenchymal and endothelial stem/progenitor cells in human postnatal life is debated. Here, we hypothesized that such progenitors are present within the human term placenta. From a heterogeneous placental single-cell suspension, a directly flow-sorted CD45-CD34+CD144+CD31Lo population uniquely differentiated into both endothelial and mesenchymal colonies in limiting dilution culture assays. Of interest, these bipotent cells were in vessel walls but not in contact with the circulation. RNA sequencing and functional analysis demonstrated that Notch signaling was a key driver for endothelial and bipotential progenitor function. In contrast, the formation of mesenchymal cells from the bipotential population was not affected by TGFß receptor inhibition, a classical pathway for endothelial-mesenchymal transition. This study reveals a bipotent progenitor phenotype in the human placenta at the cellular and molecular levels, giving rise to endothelial and mesenchymal cells ex vivo.


Asunto(s)
Células Progenitoras Endoteliales/citología , Placenta/citología , Antígenos CD/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Placenta/metabolismo , Embarazo , Receptores Notch/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología
5.
Sci Rep ; 7(1): 13558, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29051567

RESUMEN

The clinical use of endothelial colony forming cells (ECFC) is hampered by their restricted engraftment. We aimed to assess engraftment, vasculogenic and pro-angiogenic activities of ECFC in immunocompetent (C57BL/6: WT) or immunodeficient (rag1 -/- C57BL/6: Rag1) mice. In addition, the impact of host immune system was investigated where ECFC were co-implanted with mesenchymal stem/stromal cells (MSC) from adult bone marrow (AdBM-MSC), fetal bone marrow (fBM-MSC), fetal placental (fPL-MSC), or maternal placental (MPL-MSC). Transplantation of ECFCs in Matrigel plugs resulted in less cell engraftment in WT mice compared to Rag1 mice. Co-implantation with different MSCs resulted in a significant increase in cell engraftment up to 9 fold in WT mice reaching levels of engraftment observed when using ECFCs alone in Rag1 mice but well below levels of engraftment with MSC-ECFC combination in Rag1 recipients. Furthermore, MSCs did not reduce murine splenic T cell proliferation in response to ECFCs in vitro. ECFCs enhanced the murine neo-vascularization through paracrine effect, but with no difference between Rag1 and WT mice. In conclusions, the host adaptive immune system affects the engraftment of ECFCs. MSC co-implantation improves ECFC engraftment and function even in immunocompetent hosts mostly through non-immune mechanisms.


Asunto(s)
Células Endoteliales/trasplante , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Sistema Inmunológico/metabolismo , Huésped Inmunocomprometido , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina , Placenta/citología , Embarazo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Biotechnol J ; 12(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28862380

RESUMEN

Tissue engineered constructs built with human cells capable of generating a bone-like organ within the mouse have attracted considerable interest over the past decade. Here, we aimed to compare the utility of human mesenchymal stem/stromal cells (MSC) isolated from fetal term placenta (fPL-MSC) and fetal first trimester bone marrow (fBM-MSC) in a polycaprolactone scaffold/BMP7-based model in nude mice. Furthermore, fPL-MSC were co-seeded with fetal placenta-derived endothelial colony forming cells (ECFC) to assess the impact of ECFC on fPL-MSC osteogenesis. X-ray radiography and micro computed tomography analyses showed enhanced bone formation in all BMP7 groups; however there was no difference after 2 months in bone formation between scaffolds seeded with fPL-MSC alone or combination of ECFC and fPL-MSC. Of interest, fBM-MSC showed the highest level of bone formation. Additionally, endochondral ossification contributed in generation of bone in fBM-MSC. Histological analysis showed the primary role of BMP in generation of cortical and trabecular bone, and the recruitment of hematopoietic cells to the scaffolds. Current in vivo engineered bone organs can potentially be used for drug screening or as models to study bone tissue development in combination with haematopoiesis.


Asunto(s)
Células de la Médula Ósea/citología , Proteína Morfogenética Ósea 7/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Andamios del Tejido/química , Animales , Sustitutos de Huesos , Huesos/química , Huesos/citología , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Desnudos , Placenta/citología , Embarazo , Ingeniería de Tejidos
7.
Stem Cells Transl Med ; 6(4): 1070-1084, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28205414

RESUMEN

Human placenta is rich in mesenchymal stem/stromal cells (MSC), with their origin widely presumed fetal. Cultured placental MSCs are confounded by a high frequency of maternal cell contamination. Our recent systematic review concluded that only a small minority of placental MSC publications report fetal/maternal origin, and failed to discern a specific methodology for isolation of fetal MSC from term villi. We determined isolation conditions to yield fetal and separately maternal MSC during ex vivo expansion from human term placenta. MSCs were isolated via a range of methods in combination; selection from various chorionic regions, different commercial media, mononuclear cell digest and/or explant culture. Fetal and maternal cell identities were quantitated in gender-discordant pregnancies by XY chromosome fluorescence in situ hybridization. We first demonstrated reproducible maternal cell contamination in MSC cultures from all chorionic anatomical locations tested. Cultures in standard media rapidly became composed entirely of maternal cells despite isolation from fetal villi. To isolate pure fetal cells, we validated a novel isolation procedure comprising focal dissection from the cotyledonary core, collagenase/dispase digestion and explant culture in endothelial growth media that selected, and provided a proliferative environment, for fetal MSC. Comparison of MSC populations within the same placenta confirmed fetal to be smaller, more osteogenic and proliferative than maternal MSC. We conclude that in standard media, fetal chorionic villi-derived MSC (CV-MSC) do not grow readily, whereas maternal MSC proliferate to result in maternal overgrowth during culture. Instead, fetal CV-MSCs require isolation under specific conditions, which has implications for clinical trials using placental MSC. Stem Cells Translational Medicine 2017;6:1070-1084.


Asunto(s)
Vellosidades Coriónicas , Células Madre Fetales/citología , Células Madre Mesenquimatosas/citología , Placenta/citología , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Embarazo
8.
FASEB J ; 31(2): 610-624, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28045376

RESUMEN

The prospect of using endothelial progenitors is currently hampered by their low engraftment upon transplantation. We report that mesenchymal stem/stromal cells (MSCs), independent of source and age, improve the engraftment of endothelial colony forming cells (ECFCs). MSC coculture altered ECFC appearance to an elongated mesenchymal morphology with reduced proliferation. ECFC primed via MSC contact had reduced self-renewal potential, but improved capacity to form tube structures in vitro and engraftment in vivo Primed ECFCs displayed major differences in transcriptome compared to ECFCs never exposed to MSCs, affecting genes involved in the cell cycle, up-regulating of genes influencing mesenchymal transition, adhesion, extracellular matrix. Inhibition of NOTCH signaling, a potential upstream regulator of mesenchymal transition, in large part modulated this gene expression pattern and functionally reversed the mesenchymal morphology of ECFCs. The collective results showed that primed ECFCs survive better and undergo a mesenchymal transition that is dependent on NOTCH signaling, resulting in significantly increased vasculogenic potential.-Shafiee, A., Patel, J., Wong, H. Y., Donovan, P., Hutmacher, D. W., Fisk, N. M., Khosrotehrani, K. Priming of endothelial colony-forming cells in a mesenchymal niche improves engraftment and vasculogenic potential by initiating mesenchymal transition orchestrated by NOTCH signaling.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Técnicas de Cocultivo , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Placenta , Embarazo , Receptores Notch/genética
9.
Circulation ; 135(8): 786-805, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-27899395

RESUMEN

BACKGROUND: During adult life, blood vessel formation is thought to occur via angiogenic processes involving branching from existing vessels. An alternate proposal suggests that neovessels form from endothelial progenitors able to assemble the intimal layers. We here aimed to define vessel-resident endothelial progenitors in vivo in a variety of tissues in physiological and pathological situations such as normal aorta, lungs, and wound healing, tumors, and placenta, as well. METHODS: Based on protein expression levels of common endothelial markers using flow cytometry, 3 subpopulations of endothelial cells could be identified among VE-Cadherin+ and CD45- cells. RESULTS: Lineage tracing by using Cdh5creERt2/Rosa-YFP reporter strategy demonstrated that the CD31-/loVEGFR2lo/intracellular endothelial population was indeed an endovascular progenitor (EVP) of an intermediate CD31intVEGFR2lo/intracellular transit amplifying (TA) and a definitive differentiated (D) CD31hiVEGFR2hi/extracellular population. EVP cells arose from vascular-resident beds that could not be transferred by bone marrow transplantation. Furthermore, EVP displayed progenitor-like status with a high proportion of cells in a quiescent cell cycle phase as assessed in wounds, tumors, and aorta. Only EVP cells and not TA and D cells had self-renewal capacity as demonstrated by colony-forming capacity in limiting dilution and by transplantation in Matrigel plugs in recipient mice. RNA sequencing revealed prominent gene expression differences between EVP and D cells. In particular, EVP cells highly expressed genes related to progenitor function including Sox9, Il33, Egfr, and Pdfgrα. Conversely, D cells highly expressed genes related to differentiated endothelium including Ets1&2, Gata2, Cd31, Vwf, and Notch. The RNA sequencing also pointed to an essential role of the Sox18 transcription factor. The role of SOX18 in the differentiation process was validated by using lineage-tracing experiments based on Sox18CreERt2/Rosa-YFP mice. Besides, in the absence of functional SOX18/SOXF, EVP progenitors were still present, but TA and D populations were significantly reduced. CONCLUSIONS: Our findings support an entirely novel endothelial hierarchy, from EVP to TA to D, as defined by self-renewal, differentiation, and molecular profiling of an endothelial progenitor. This paradigm shift in our understanding of vascular-resident endothelial progenitors in tissue regeneration opens new avenues for better understanding of cardiovascular biology.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre/metabolismo , Animales , Antígenos CD/metabolismo , Aorta/metabolismo , Aorta/patología , Trasplante de Médula Ósea , Cadherinas/metabolismo , Diferenciación Celular , Células Endoteliales/citología , Endotelio Vascular/citología , Femenino , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Placenta/metabolismo , Placenta/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Factores de Transcripción SOXF/metabolismo , Células Madre/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Heridas y Lesiones/patología , Heridas y Lesiones/terapia
10.
PeerJ ; 4: e1845, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27042394

RESUMEN

Mesenchymal stromal cells (MSC) are widely used for the study of mesenchymal tissue repair, and increasingly adopted for cell therapy, despite the lack of consensus on the identity of these cells. In part this is due to the lack of specificity of MSC markers. Distinguishing MSC from other stromal cells such as fibroblasts is particularly difficult using standard analysis of surface proteins, and there is an urgent need for improved classification approaches. Transcriptome profiling is commonly used to describe and compare different cell types; however, efforts to identify specific markers of rare cellular subsets may be confounded by the small sample sizes of most studies. Consequently, it is difficult to derive reproducible, and therefore useful markers. We addressed the question of MSC classification with a large integrative analysis of many public MSC datasets. We derived a sparse classifier (The Rohart MSC test) that accurately distinguished MSC from non-MSC samples with >97% accuracy on an internal training set of 635 samples from 41 studies derived on 10 different microarray platforms. The classifier was validated on an external test set of 1,291 samples from 65 studies derived on 15 different platforms, with >95% accuracy. The genes that contribute to the MSC classifier formed a protein-interaction network that included known MSC markers. Further evidence of the relevance of this new MSC panel came from the high number of Mendelian disorders associated with mutations in more than 65% of the network. These result in mesenchymal defects, particularly impacting on skeletal growth and function. The Rohart MSC test is a simple in silico test that accurately discriminates MSC from fibroblasts, other adult stem/progenitor cell types or differentiated stromal cells. It has been implemented in the www.stemformatics.org resource, to assist researchers wishing to benchmark their own MSC datasets or data from the public domain. The code is available from the CRAN repository and all data used to generate the MSC test is available to download via the Gene Expression Omnibus or the Stemformatics resource.

11.
Stem Cells ; 34(4): 902-12, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26732848

RESUMEN

Since the discovery of endothelial colony forming cells (ECFC), there has been significant interest in their therapeutic potential to treat vascular injuries. ECFC cultures display significant heterogeneity and a hierarchy among cells able to give rise to high proliferative versus low proliferative colonies. Here we aimed to define molecularly this in vitro hierarchy. Based on flow cytometry, CD34 expression levels distinguished two populations. Only CD34 + ECFC had the capacity to reproduce high proliferative potential (HPP) colonies on replating, whereas CD34- ECFCs formed only small clusters. CD34 + ECFCs were the only ones to self-renew in stringent single-cell cultures and gave rise to both CD34 + and CD34- cells. Upon replating, CD34 + ECFCs were always found at the centre of HPP colonies and were more likely in G0/1 phase of cell cycling. Functionally, CD34 + ECFC were superior at restoring perfusion and better engrafted when injected into ischemic hind limbs. Transcriptomic analysis identified cyclin-dependent kinase (CDK) cell cycle inhibiting genes (p16, p21, and p57), the Notch signaling pathway (dll1, dll4, hes1, and hey1), and the endothelial cytokine il33 as highly expressed in CD34 + ECFC. Blocking the Notch pathway using a γ-secretase inhibitor (DAPT) led to reduced expression of cell cycle inhibitors, increased cell proliferation followed by a loss of self-renewal, and HPP colony formation capacity reflecting progenitor exhaustion. Similarly shRNA knockdown of p57 strongly affected self-renewal of ECFC colonies. ECFC hierarchy is defined by Notch signalling driving cell cycle regulators, progenitor quiescence and self-renewal potential.


Asunto(s)
Antígenos CD34/metabolismo , Células Progenitoras Endoteliales/trasplante , Neovascularización Fisiológica/genética , Receptores Notch/genética , Lesiones del Sistema Vascular/terapia , Animales , Linaje de la Célula/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Dipéptidos/administración & dosificación , Células Progenitoras Endoteliales/metabolismo , Citometría de Flujo , Miembro Posterior/patología , Miembro Posterior/trasplante , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Receptores Notch/antagonistas & inhibidores , Receptores Notch/biosíntesis , Medicina Regenerativa , Transducción de Señal/efectos de los fármacos , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
12.
Stem Cells Dev ; 25(5): 395-404, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26728561

RESUMEN

Alport syndrome (AS) is a hereditary glomerulopathy caused by a mutation in type IV collagen genes, which disrupts glomerular basement membrane, leading to progressive glomerulosclerosis and end-stage renal failure. There is at present no cure for AS, and cell-based therapies offer promise to improve renal function. In this study, we found that human first trimester fetal chorionic stem cells (CSC) are able to migrate to glomeruli and differentiate down the podocyte lineage in vitro and in vivo. When transplanted into 7-week-old Alport 129Sv-Col4α3(tm1Dec)/J (-/-) mice, a single intraperitoneal injection of CSC significantly lowered blood urea and urine proteinuria levels over the ensuing 2 weeks. In addition, nearly two-thirds of transplanted -/- mice maintained their weight above the 80% welfare threshold, with both males and females weighing more than age-matched nontransplanted -/- mice. This was associated with less renal cortical fibrosis and interstitial inflammation compared to nontransplanted mice as shown by reduction in murine CD4, CD68, and CD45.2 cells. Transplanted CSC homed to glomeruli, where they expressed CR1, VEGFA, SYNAPTOPODIN, CD2AP, and PODOCIN at the RNA level and produced PODOCIN, CD2AP, and COLIVα3 proteins in nontransplanted -/- mice, indicating that CSC have adopted a podocyte phenotype. Together, these data indicate that CSC may be used to delay progression of renal pathology by a combination of anti-inflammatory effects and replacement of the defective resident podocytes.


Asunto(s)
Diferenciación Celular , Corion/citología , Nefritis Hereditaria/terapia , Podocitos/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Corion/trasplante , Técnicas de Cocultivo , Colágeno Tipo IV/farmacología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fibrosis , Humanos , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Corteza Renal/patología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Mutación/genética , Nefritis Hereditaria/patología , Fenotipo , Podocitos/efectos de los fármacos , Podocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
13.
Org Lett ; 17(24): 5998-6001, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26629727

RESUMEN

An approach to the dihydrooxepino[4,3-b]pyrrole core of diketopiperazine natural products which utilizes a vinyl pyrrole epoxide Cope rearrangement was investigated. It was found that an ester substituent on the epoxide was essential for the [3,3]-rearrangement to occur. Density functional calculations with M06-2X provided explanations for the effects of the pyrrole and ester groups on these rearrangements.


Asunto(s)
Productos Biológicos/síntesis química , Dicetopiperazinas/química , Compuestos Epoxi/síntesis química , Pirroles/síntesis química , Productos Biológicos/química , Compuestos Epoxi/química , Ésteres , Estructura Molecular , Piperazinas/química , Piperazinas/aislamiento & purificación , Pirroles/química , Relación Estructura-Actividad
14.
Stem Cell Reports ; 5(5): 682-689, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26527386

RESUMEN

In this article, Millard and colleagues show that intrauterine bone marrow transplantation in the oim/oim mouse model of osteogenesis imperfecta yields hematopoietic microchimerism in the absence of donor osteopoiesis or phenotypic improvement. Bone-associated donor cells were not bone-forming osteoblasts, but osteoclasts (bone resorbing cells of the hematopoietic lineage) and osteal macrophages (bone regulatory cells of the hematopoietic lineage).


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Osteoblastos/citología , Osteoclastos/citología , Osteogénesis Imperfecta/cirugía , Animales , Ratones
15.
Endocrine ; 49(3): 643-52, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26067082

RESUMEN

Analysis of archival samples from cohorts of pregnant women may be key to discovering prognosticators of stillbirth and pregnancy/perinatal complications. Growth hormone (GH) and its receptor (GHR) are pivotal in feto-placental development and pregnancy maintenance. We report a rapid, optimized method for genotyping the GHR full-length versus exon 3-deleted isoform (GHRd3). TaqMan single nucleotide polymorphism (SNP) genotyping proved superior to standard multiplex polymerase chain reaction (PCR) in allele detection and GHR genotyping from archived samples, including those with poor genomic deoxyribonucleic acid quality/quantity such as formalin fixed, paraffin embedded, blood, and serum. Furthermore, this assay is suitable for high through put 96 or 384-well plate quantitative PCR machines with automated genotype calling software. The TaqMan genotyping assay can increase the data obtained from precious archival human samples.


Asunto(s)
Proteínas Portadoras/genética , Placenta/química , Adulto , Australia/epidemiología , Células Cultivadas , Estudios de Cohortes , Exones , Femenino , Sangre Fetal/química , Eliminación de Gen , Frecuencia de los Genes , Genotipo , Humanos , Desequilibrio de Ligamiento , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética , Embarazo , Fijación del Tejido
16.
Stem Cells Transl Med ; 4(5): 419-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25769652

RESUMEN

Since the isolation of fetal stem cell populations from perinatal tissues, such as umbilical cord blood and placenta, interest has been growing in understanding their greater plasticity compared with adult stem cells and exploring their potential in regenerative medicine. The phenomenon of fetal microchimerism (FMC) naturally occurring during pregnancy through the transfer of fetal stem/progenitor cells to maternal blood and tissues has been integral in developing this dogma. Specifically, microchimeric mesenchymal stem cells and endothelial progenitors of fetal origin have now demonstrated a capacity for tissue repair in the maternal host. However, the use of similar fetal stem cells in therapy has been significantly hampered by the availability of clinically relevant cell numbers and/or contamination with cells of maternal origin, particularly when using the chorionic and decidual placenta. In the present prospective review, we highlight the importance of FMC to the field of fetal stem cell biology and issues of maternal contamination from perinatal tissues and discuss specific isolation strategies to overcome these translational obstacles.


Asunto(s)
Células Madre Fetales/citología , Células Madre Mesenquimatosas/citología , Placenta/citología , Medicina Regenerativa , Adulto , Diferenciación Celular/genética , Quimerismo , Femenino , Humanos , Embarazo
17.
Stem Cells Transl Med ; 3(11): 1305-11, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25154781

RESUMEN

Placenta is a readily accessible translationally advantageous source of mesenchymal stem/stromal cells (MSCs) currently used in cryobanking and clinical trials. MSCs cultured from human chorion have been widely assumed to be fetal in origin, despite evidence that placental MSCs may be contaminated with maternal cells, resulting in entirely maternally derived MSC cultures. To document the frequency and determinants of maternal cell contamination in chorionic MSCs, we undertook a PRISMA-compliant systematic review of publications in the PubMed, Medline, and Embase databases (January 2000 to July 2013) on placental and/or chorionic MSCs from uncomplicated pregnancies. Of 147 studies, only 26 (18%) investigated fetal and/or maternal cell origin. After excluding studies that did not satisfy minimal MSC criteria, 7 of 15 informative studies documented MSC cultures as entirely fetal, a further 7 studies reported cultured human chorionic MSC populations to be either maternal (n=6) or mixed (n=1), whereas 1 study separately cultured pure fetal and pure maternal MSC from the same placenta. Maternal cell contamination was associated with term and chorionic membrane samples and greater passage number but was still present in 30% of studies of chorionic villous MSCs. Although most studies assume fetal origin for MSCs sourced from chorion, this systematic review documents a high incidence of maternal-origin MSC populations in placental MSC cultures. Given that fetal MSCs have more primitive properties than adult MSCs, our findings have implications for clinical trials in which knowledge of donor and tissue source is pivotal. We recommend sensitive methods to quantitate the source and purity of placental MSCs.


Asunto(s)
Células Madre Adultas , Corion , Células Madre Mesenquimatosas , Placenta , Adulto , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Corion/citología , Corion/metabolismo , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Placenta/citología , Placenta/metabolismo , Embarazo
18.
BMC Cell Biol ; 15: 15, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24885150

RESUMEN

BACKGROUND: Fetal mesenchymal stem/stromal cells (MSC) represent a developmentally-advantageous cell type with translational potential.To enhance adult MSC migration, studies have focussed on the role of the chemokine receptor CXCR4 and its ligand SDF-1 (CXCL12), but more recent work implicates an intricate system of CXCR4 receptor dimerization, intracellular localization, multiple ligands, splice variants and nuclear accumulation. We investigated the intracellular localization of CXCR4 in fetal bone marrow-derived MSC and role of intracellular trafficking in CXCR4 surface expression and function. RESULTS: We found that up to 4% of human fetal MSC have detectable surface-localized CXCR4. In the majority of cells, CXCR4 is located not at the cell surface, as would be required for 'sensing' migratory cues, but intracellularly. CXCR4 was identified in early endosomes, recycling endosomes, and lysosomes, indicating only a small percentage of CXCR4 travelling to the plasma membrane. Notably CXCR4 was also found in and around the nucleus, as detected with an anti-CXCR4 antibody directed specifically against CXCR4 isoform 2 differing only in N-terminal sequence. After demonstrating that endocytosis of CXCR4 is largely independent of endogenously-produced SDF-1, we next applied the cytoskeletal inhibitors blebbistatin and dynasore to inhibit endocytotic recycling. These increased the number of cells expressing surface CXCR4 by 10 and 5 fold respectively, and enhanced the number of cells migrating to SDF1 in vitro (up to 2.6 fold). These molecules had a transient effect on cell morphology and adhesion, which abated after the removal of the inhibitors, and did not alter functional stem cell properties. CONCLUSIONS: We conclude that constitutive endocytosis is implicated in the regulation of CXCR4 membrane expression, and suggest a novel pharmacological strategy to enhance migration of systemically-transplanted cells.


Asunto(s)
Endocitosis , Feto/citología , Células Madre Mesenquimatosas/citología , Receptores CXCR4/análisis , Receptores CXCR4/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Movimiento Celular , Núcleo Celular/metabolismo , Células Cultivadas , Endosomas/metabolismo , Femenino , Humanos , Lisosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transporte de Proteínas
19.
Stem Cells Dev ; 23(3): 262-76, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24028330

RESUMEN

Osteogenesis imperfecta (OI) is a genetic bone pathology with prenatal onset, characterized by brittle bones in response to abnormal collagen composition. There is presently no cure for OI. We previously showed that human first trimester fetal blood mesenchymal stem cells (MSCs) transplanted into a murine OI model (oim mice) improved the phenotype. However, the clinical use of fetal MSC is constrained by their limited number and low availability. In contrast, human fetal early chorionic stem cells (e-CSC) can be used without ethical restrictions and isolated in high numbers from the placenta during ongoing pregnancy. Here, we show that intraperitoneal injection of e-CSC in oim neonates reduced fractures, increased bone ductility and bone volume (BV), increased the numbers of hypertrophic chondrocytes, and upregulated endogenous genes involved in endochondral and intramembranous ossification. Exogenous cells preferentially homed to long bone epiphyses, expressed osteoblast genes, and produced collagen COL1A2. Together, our data suggest that exogenous cells decrease bone brittleness and BV by directly differentiating to osteoblasts and indirectly stimulating host chondrogenesis and osteogenesis. In conclusion, the placenta is a practical source of stem cells for the treatment of OI.


Asunto(s)
Corion/citología , Células Madre Fetales/citología , Células Madre Fetales/trasplante , Fracturas Óseas/terapia , Osteogénesis Imperfecta/terapia , Placenta/citología , Animales , Huesos/anomalías , Huesos/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis , Corion/metabolismo , Colágeno Tipo I/agonistas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Madre Fetales/metabolismo , Feto , Fracturas Óseas/genética , Fracturas Óseas/metabolismo , Fracturas Óseas/patología , Expresión Génica , Humanos , Inyecciones Intraperitoneales , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Placenta/metabolismo , Embarazo , Trasplante de Células Madre , Trasplante Heterólogo
20.
Kidney Int ; 85(3): 600-10, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24304884

RESUMEN

Fetal microchimeric cells (FMCs) enter the maternal circulation and persist in tissue for decades. They have capacity to home to injured maternal tissue and differentiate along that tissue's lineage. This raises the question of the origin(s) of cells transferred to the mother during pregnancy. FMCs with a mesenchymal phenotype have been documented in several studies, which makes mesenchymal stem cells an attractive explanation for their broad plasticity. Here we assessed the recruitment and mesenchymal lineage contribution of FMCs in response to acute kidney fibrosis induced by aristolochic acid injection. Serial in vivo bioluminescence imaging revealed a biphasic recruitment of active collagen-producing FMCs during the repair process of injured kidney in post-partum wild-type mothers that had delivered transgenic pups expressing luciferase under the collagen type I-promoter. The presence of FMCs long-term post injury (day 60) was associated with profibrotic molecules (TGF-ß/CTGF), serum urea levels, and collagen deposition. Immunostaining confirmed FMCs at short term (day 15) using post-partum wild-type mothers that had delivered green fluorescent protein-positive pups and suggested a mainly hematopoietic phenotype. We conclude that there is biphasic recruitment to, and activity of, FMCs at the injury site. Moreover, we identified five types of FMC, implicating them all in the reparative process at different stages of induced renal interstitial fibrosis.


Asunto(s)
Lesión Renal Aguda/patología , Quimerismo/embriología , Feto/citología , Riñón/patología , Animales , Movimiento Celular , Femenino , Fibrosis , Hematopoyesis , Células Madre Mesenquimatosas/fisiología , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA