Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biomed Mater Res A ; 112(9): 1484-1493, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38487991

RESUMEN

Mesenchymal stem cell-derived secretome represents an emerging acellular therapeutic which possess significant opportunity for clinical applications due to its anti-inflammatory, immunomodulatory, and wound healing properties. However, maintaining therapeutic efficacy and ensuring stability of cell-based products is challenging, requiring a robust delivery method. Therefore, we designed a hydrogel-based scaffold loaded with CK Cell Technologies' proprietary Mesenchymal stem cell-secretome for controlled release treatment of acute and chronic wounds. We incorporated both conditioned media (CM) and extracellular vesicles (EVs) into gelatin methacryloyl (GelMA) hydrogels and demonstrated how we can tune the diffusive release of the EVs from them. To demonstrate viability of the approach, we developed a wound healing scratch assay where we see in situ release of CM and EVs promote enhanced migration of human dermal fibroblasts (hDFs). We see the colocalization of these EVs in the fibroblasts using fluorescent microscopy. Finally, as a surrogate for in vivo neovascularization, we conducted an in vitro tube formation assay for the MSC-secretome using matrigel-embedded human microvascular endothelial cells. By adding CM and EVs, we observe an increase in tubulogenesis. Collectively, our data demonstrates by tuning the GelMA properties, we can influence the controlled release of the MSC-secretome for a wound dressing and bandage application for chronic and acute wounds.


Asunto(s)
Gelatina , Hidrogeles , Células Madre Mesenquimatosas , Cicatrización de Heridas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Humanos , Hidrogeles/química , Cicatrización de Heridas/efectos de los fármacos , Gelatina/química , Fibroblastos/citología , Vesículas Extracelulares/metabolismo , Polimerizacion , Metacrilatos/química , Movimiento Celular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología
3.
J Stem Cells Regen Med ; 18(1): 2-10, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36003657

RESUMEN

Mesenchymal stem cells derived from adipose tissue (ADMSCs) are being increasingly considered in regenerative medicine-based clinical applications. Apart from possessing therapeutic applications themselves, ADMSCs also secrete a myriad of soluble factors which are promising candidates for treating several degenerative diseases such as osteoarthritis and neurodegenerative diseases, wound repair as well as for cosmeceutical purposes. In our research study, we successfully isolated ADMSCs in-house, now called CKC-Endeavour-1 from the lipoaspirate sample of a patient who underwent liposuction. The subsequent expansion of cells was performed in xeno-free and serum-free conditions and their characterisation was performed using tri-lineage differentiation studies. The levels of differentiation were assessed by staining and gene expression which was observed to be comparable between the in-house developed ADMSC cell line and the commercially purchased ADMSCs. Following characterisation, the secretory components from these MSCs, namely, conditioned media (ADMSC-CM) and exosomes (ADMSC-EXO) were harvested from CKC-Endeavour-1 under xeno-free, serum-free, and supplement-free conditions followed by lyophilisation in order to attempt to prolong its shelf-life. The comprehensive analysis of the secretome profile of ADMSC-CM using carried out using cytokine array and demonstrated the presence of 105 cytokines and growth factors. Also, clinical grade Izon columns were used to isolate the exosomes from ADMSC-CM obtaining exosomes in the size range of <200nm, analysed using nanoparticle tracking analysis. Overall, our study developed an ADMSC cell line, CKC-Endeavour-1, along with their CM and exosome (EXO) products under clinically safe conditions. Additionally, we have obtained a comprehensive understanding of the secreted factors present in the ADMSC-CM which could be further explored in detail to tap the best therapeutic benefits from them.

4.
J Alzheimers Dis ; 83(4): 1563-1601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34487051

RESUMEN

Neurological disorders significantly impact the world's economy due to their often chronic and life-threatening nature afflicting individuals which, in turn, creates a global disease burden. The Group of Twenty (G20) member nations, which represent the largest economies globally, should come together to formulate a plan on how to overcome this burden. The Neuroscience-20 (N20) initiative of the Society for Brain Mapping and Therapeutics (SBMT) is at the vanguard of this global collaboration to comprehensively raise awareness about brain, spine, and mental disorders worldwide. This paper aims to provide a comprehensive review of the various brain initiatives worldwide and highlight the need for cooperation and recommend ways to bring down costs associated with the discovery and treatment of neurological disorders. Our systematic search revealed that the cost of neurological and psychiatric disorders to the world economy by 2030 is roughly $16T. The cost to the economy of the United States is $1.5T annually and growing given the impact of COVID-19. We also discovered there is a shortfall of effective collaboration between nations and a lack of resources in developing countries. Current statistical analyses on the cost of neurological disorders to the world economy strongly suggest that there is a great need for investment in neurotechnology and innovation or fast-tracking therapeutics and diagnostics to curb these costs. During the current COVID-19 pandemic, SBMT, through this paper, intends to showcase the importance of worldwide collaborations to reduce the population's economic and health burden, specifically regarding neurological/brain, spine, and mental disorders.


Asunto(s)
Carga Global de Enfermedades , Cooperación Internacional , Trastornos Mentales , Enfermedades del Sistema Nervioso , COVID-19/epidemiología , Carga Global de Enfermedades/organización & administración , Carga Global de Enfermedades/tendencias , Salud Global/economía , Salud Global/tendencias , Humanos , Trastornos Mentales/economía , Trastornos Mentales/epidemiología , Trastornos Mentales/terapia , Enfermedades del Sistema Nervioso/economía , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/terapia , Neurociencias/métodos , Neurociencias/tendencias , SARS-CoV-2
5.
J Cell Biochem ; 120(2): 1174-1184, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30335884

RESUMEN

Reelin plays important roles in brain development. Reeler mutant mice that lack the protein reelin (RELN) suffer from cell type- and region-dependent changes in their neocortical layers, and adult reeler mutant mice have dilated seminiferous tubules. Meanwhile, the mechanism by which Reelin regulates the spermatogenic cell development in mice and their reproductive abilities remains unclear. In the present study, we used reeler mutant mice to investigate the effects of Reelin on reproduction in mice. The results indicated variations in sex hormone expression among the reeler mice, indicating that they produce few offspring and their spermatogenic cells are irregularly developed. Moreover, glial cell line-derived neurotrophic factor (GDNF)/GDNF family receptor alpha 1, Ras/extracellular regulated protein kinases (ERK), and promyelocytic leukemia zinc finger (PLZF)/chemokine (C-X-C motif) receptor 4 (CXCR4) serve as potential regulatory pathways that respond to the changes in sertoli cells and the niche of male germ cells. Our findings provided valuable insights into the role of reeler in the reproductive abilities of male mice and development of their spermatogonia stem cells.

6.
Stem Cell Res ; 32: 135-138, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30278375

RESUMEN

Peripheral dermal fibroblasts (DF) from a healthy 56 year old female were obtained from the Centre for Healthy Brain Ageing (CHeBA) Biobank, University of New South Wales, under the material transfer agreement with the University of Wollongong. DFs were reprogrammed via mRNA-delivered transcription factors into induced pluripotent stem cells (iPSCs). The generated iPSCs were confirmed to be pluripotent, capable of three germ layer differentiation and are thus a useful resource for creating iPSC-derived healthy human cells of any lineage. Resource table.


Asunto(s)
Reprogramación Celular/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Piel/citología , Células Cultivadas , Reprogramación Celular/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Persona de Mediana Edad
7.
Cell Death Dis ; 9(10): 968, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237484

RESUMEN

Diabetes mellitus affects a large number of men of reproductive age and it usually leads to serious reproductive disorders. However, the underlying mechanisms and specific therapies still remain largely unknown. We observed Leydig cell loss in the testes of diabetic mice. Continuous high glycemic status of testes stimulated expression of Caspase12, Grp78, and Chop, the three ERS response factors; this might induce cell cycle arrest and apoptosis of Leydig cells in response to ERS. In these diabetic mouse models, melatonin alleviated apoptosis of testicular stromal cell induced by ERS, and promoted SSCs self-renewal by recovering Leydig cells secretion of CSF1 after 8 weeks of treatment. To explore the relationship between CSF-1 and ERS in Leydig cells, we treated Leydig tumor cell line with an activator Tuniamycin and an inhibitor 4-Phenylbutyrate of ERS. Our data showed that the CSF-1 expression in mouse Leydig cell lines decreased six-fold while reversely increasing five-fold in the 4-Phenylbutyrate-treated group. Thus, melatonin likely alleviates the loss of Leydig cells in diabetic testes and provides a healthier niche for SSCs to self-renew and continually provide healthy sperm for male fertility.


Asunto(s)
Células Intersticiales del Testículo/efectos de los fármacos , Melatonina/uso terapéutico , Espermatogonias/efectos de los fármacos , Células Madre Germinales Adultas/citología , Células Madre Germinales Adultas/efectos de los fármacos , Células Madre Germinales Adultas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Espermatogonias/citología , Espermatogonias/metabolismo , Testículo/citología , Testículo/efectos de los fármacos , Testículo/metabolismo
8.
J Cell Physiol ; 234(1): 915-926, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30069947

RESUMEN

Self-renewal and differentiation of male germline stem cells (mGSCs) provide the basic function for continual spermatogenesis. Studies of in vitro culture of germline stem cells are important and meaningful for basic biological research and practical application. Growth factors, such as GDNF, bFGF, CSF1, and EGF, could maintain the self-renewal of mGSCs. Insulin-like growth factor 1 (IGF-1), an important growth factor, and its pathway have been reported to maintain the survival of several types of stem cells and play important roles in male reproduction. However, the mechanism through which the IGF-1 pathway acts to regulate the self-renewal of mGSCs remains unclear. We analyzed the effect of IGF-1 on the proliferation and apoptosis of bovine mGSCs. We evaluated the expression profile of long noncoding RNA (LncRNA) H19 in bovine and mouse tissues. Moreover, we investigated whether LncRNA H19 could regulate the IGF-1 pathway. Results showed that IGF-1 could activate the phosphorylation of AKT and ERK signaling pathways, and the IGF-1 pathway played an important role in regulating the proliferation and apoptosis of bovine mGSCs. The proliferation rate of mGSCs decreased, whereas the apoptosis rate of mGSCs increased when the IGF-1 receptor (IGF-1R) was blocked using the IGF-1R-specific inhibitor (picropodophyllin). LncRNA H19 could regulate the IGF-1 signaling pathway and, consequently, the proliferation and apoptosis of mGSCs. The number of cells in the seminiferous tubule decreased when H19 was interfered by injecting a virus-containing supernatant. Hence, LncRNA H19 participated in the regulation of the proliferation and apoptosis of mGSCs via the IGF-1 signaling pathway.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/genética , ARN Largo no Codificante/genética , Espermatogénesis/genética , Células Madre/citología , Animales , Apoptosis/genética , Bovinos , Diferenciación Celular/genética , Proliferación Celular/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Ratones , Transducción de Señal/genética , Células Madre/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
9.
Stem Cell Reports ; 9(2): 557-570, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28793248

RESUMEN

Repopulation of brain circuits by neural precursors is a potential therapeutic strategy for neurodegenerative disorders; however, choice of cell is critical. Previously, we introduced a two-step culture system that generates a high yield of neural precursors from small samples of adult canine skin. Here, we probe their gene and protein expression profiles in comparison with dermal fibroblasts and brain-derived neural stem cells and characterize their neuronal potential. To date, we have produced >50 skin-derived neural precursor (SKN) lines. SKNs can be cultured in a highly replicable fashion and uniformly express a panel of identifying markers. Upon differentiation, they self-upregulate neural specification genes, generating neurons with basic electrophysiological functionality. This unique population of neural precursors, derived from mature skin, overcomes many of the practical issues that have limited clinical translation of alternative cell types. Easily accessible, neuronally committed, and patient specific, SKNs may have potential for the treatment of brain disorders.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Piel/citología , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Proliferación Celular , Células Cultivadas , Perros , Perfilación de la Expresión Génica , Inmunofenotipificación , Fenotipo , Sinapsis/metabolismo
10.
Sci Rep ; 6: 31450, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27514990

RESUMEN

Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, yet current therapeutic treatments are inadequate due to a complex disease pathogenesis. The plant polyphenol apigenin has been shown to have anti-inflammatory and neuroprotective properties in a number of cell and animal models; however a comprehensive assessment has not been performed in a human model of AD. Here we have used a human induced pluripotent stem cell (iPSC) model of familial and sporadic AD, in addition to healthy controls, to assess the neuroprotective activity of apigenin. The iPSC-derived AD neurons demonstrated a hyper-excitable calcium signalling phenotype, elevated levels of nitrite, increased cytotoxicity and apoptosis, reduced neurite length and increased susceptibility to inflammatory stress challenge from activated murine microglia, in comparison to control neurons. We identified that apigenin has potent anti-inflammatory properties with the ability to protect neurites and cell viability by promoting a global down-regulation of cytokine and nitric oxide (NO) release in inflammatory cells. In addition, we show that apigenin is able to protect iPSC-derived AD neurons via multiple means by reducing the frequency of spontaneous Ca(2+) signals and significantly reducing caspase-3/7 mediated apoptosis. These data demonstrate the broad neuroprotective action of apigenin against AD pathogenesis in a human disease model.


Asunto(s)
Enfermedad de Alzheimer/patología , Apigenina/farmacología , Apoptosis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Adulto , Anciano , Enfermedad de Alzheimer/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/patología , Inflamación/prevención & control , Interferón gamma/farmacología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico
11.
Neurosci Lett ; 593: 107-13, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25778416

RESUMEN

During evolution a unique anterior-posterior flexure posited the canine dentate gyrus in two distinct dorsal and ventral positions. We therefore sought to explore neurogenesis and neurogenic cell-related difference along the canine hippocampal dorsal-ventral axis. Post mortem histological analysis revealed 49.1% greater doublecortin (DCX)-positive cells and a 158.5% greater percentage of double labeled DCX-positive/neuronal nuclei (NeuN) positive cells in the dorsal subgranular zone compared to the ventral. We then show neural precursor cells isolated from fresh hippocampal tissue are capable of proliferating long term, and after differentiation, express neuronal and glial markers. Dorsal hippocampal isolates produced a 120.0% higher frequency of sphere-forming neural precursor cells compared to ventral hippocampal tissue. Histological DCX and neurosphere assay results were highly correlated. Overall, we provide the first evidence that the dorsal canine hippocampus has a markedly higher rate of adult neurogenesis than the ventral hippocampus, possibly related to a greater frequency of contributory neural precursor cells.


Asunto(s)
Perros/anatomía & histología , Hipocampo/citología , Células-Madre Neurales/citología , Neurogénesis , Animales , Diferenciación Celular , Giro Dentado/anatomía & histología , Giro Dentado/citología , Femenino , Hipocampo/anatomía & histología , Masculino , Neuroglía/citología , Neuronas/citología
12.
Stem Cell Res ; 11(3): 978-89, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23900167

RESUMEN

Human embryonic stem cells (hESCs) are emerging as an attractive alternative source for cell replacement therapy since the cells can be expanded in culture indefinitely and differentiated into any cell types in the body. In order to optimize cell-to-cell interaction, cell proliferation and differentiation into specific lineages as well as tissue organization, it is important to provide a microenvironment for the hESCs which mimics the stem cell niche. One approach is to provide a three-dimensional (3D) environment such as encapsulation. We present an approach to culture and differentiate hESCs into midbrain dopamine (mdDA) neurons in a 3D microenvironment using alginate microcapsules for the first time. A detailed gene and protein expression analysis during neuronal differentiation showed an increased gene and protein expression of various specific DA neuronal markers, particularly tyrosine hydroxylase (TH) by >100 folds after 2 weeks and at least 50% higher expression after 4 weeks respectively, compared to cells differentiated under conventional two-dimensional (2D) platform. The encapsulated TH(+) cells co-expressed mdDA neuronal markers, forkhead box protein A-2 (FOXA2) and pituitary homeobox-3 (PITX3) after 4 weeks and secreted approximately 60pg/ml/10(6) cells higher DA level when induced. We propose that the 3D platform facilitated an early onset of DA neuronal generation compared to that with conventional 2D system which also secretes more DA under potassium-induction. It is a very useful model to study the proliferation and directed differentiation of hESCs to various lineages, particularly to mdDA neurons. This 3D system also allows the separation of feeder cells from hESCs during the process of differentiation and also has potential for immune-isolation during transplantation studies.


Asunto(s)
Alginatos/química , Cápsulas/química , Neuronas Dopaminérgicas/citología , Células Madre Embrionarias/citología , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Linaje de la Célula , Proliferación Celular , Técnicas de Cocultivo , Dopamina/metabolismo , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/metabolismo , Ácido Glucurónico/química , Factor Nuclear 3-beta del Hepatocito/metabolismo , Ácidos Hexurónicos/química , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Modelos Biológicos , Neurogénesis , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
13.
J Neural Transm (Vienna) ; 120(1): 103-11, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22695755

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative brain disorder that leads to a progressive decline in a person's memory and ability to communicate and carry out daily activities. The brain pathology in AD is characterized by extensive neuronal loss, particularly of cholinergic neurons, intracellular neurofibrillary tangles composed of the tau protein (NFTs) and extracellular deposition of plaques composed of ß-amyloid (Aß), a cleavage product of the amyloid precursor protein (APP). These two insoluble protein aggregates are accompanied by a chronic inflammatory response and extensive oxidative damage. Whereas dys-regulation of APP expression or processing appears to be important for the familial, early-onset form of AD, controversy exists between the "Baptists" (in favour of Aß) and the "Tauists" (in favour of tau) as to which of these two protein dysfunctions occur at the earliest stages or are the most important contributors to the disease process in sporadic AD. However, more and more "non-amyloid" and "non-tau" causes have been proposed, including, glycation, inflammation, oxidative stress and dys-regulation of the cell cycle. However, to get an insight into the ultimate cause of AD, and to prove that any drug target is valuable in AD, disease-relevant models giving insight into the pathogenic processes in AD are urgently needed. In the absence of a good animal model for sporadic AD, we propose in this review that induced pluripotent stem cells, derived from dermal fibroblasts of AD patients, and differentiated into cholinergic neurons, might be a promising novel tool for disease modelling and drug discovery for the sporadic form of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Antiparkinsonianos/farmacología , Descubrimiento de Drogas , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Animales , Antiparkinsonianos/uso terapéutico , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas tau/metabolismo
14.
Biol Psychiatry ; 73(5): 435-42, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22795967

RESUMEN

BACKGROUND: Physical exercise in early adulthood and mid-life improves cognitive function and enhances brain plasticity, but the effects of commencing exercise in late adulthood are not well-understood. METHOD: We investigated the effects of voluntary exercise in the restoration of place recognition memory in aged rats and examined hippocampal changes of synaptic density and neurogenesis. RESULTS: We found a highly selective age-related deficit in place recognition memory that is stable across retest sessions and correlates strongly with loss of hippocampal synapses. Additionally, 12 weeks of voluntary running at 20 months of age removed the deficit in the hippocampally dependent place recognition memory. Voluntary running restored presynaptic density in the dentate gyrus and CA3 hippocampal subregions in aged rats to levels beyond those observed in younger animals, in which exercise had no functional or synaptic effects. By contrast, hippocampal neurogenesis, a possible memory-related mechanism, increased in both young and aged rats after physical exercise but was not linked with performance in the place recognition task. We used graph-based network analysis based on synaptic covariance patterns to characterize efficient intrahippocampal connectivity. This analysis revealed that voluntary running completely reverses the profound degradation of hippocampal network efficiency that accompanies sedentary aging. Furthermore, at an individual animal level, both overall hippocampal presynaptic density and subregional connectivity independently contribute to prediction of successful place recognition memory performance. CONCLUSIONS: Our findings emphasize the unique synaptic effects of exercise on the aged brain and their specific relevance to a hippocampally based memory system for place recognition.


Asunto(s)
Encéfalo/fisiología , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Red Nerviosa/fisiología , Neurogénesis/fisiología , Condicionamiento Físico Animal/psicología , Factores de Edad , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Hipocampo/metabolismo , Red Nerviosa/metabolismo , Condicionamiento Físico Animal/fisiología , Ratas , Carrera
15.
Methods Mol Biol ; 873: 237-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22528359

RESUMEN

The differentiation of human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) towards functional neurons particularly hold great potential for the cell-based replacement therapy in neurodegenerative diseases. Here, we describe a stepwise differentiation protocol that mimics the early stage of neural development in human to promote the generation of neuroprogenitors at a high yield. Both the hESCs and hiPSCs are initially cultured in an optimized feeder-free condition, which offer an efficient formation of aggregates. To specify the neuroectodermal specification, these aggregates are differentiated in a defined neural induction medium to develop into neural rosettes-like structures. The rosettes are expanded into free-floating sphere and can be further propagated or developed into variety of neuronal subtypes.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Neuronas/citología , Células Madre Pluripotentes/citología , Células Madre/citología , Diferenciación Celular/fisiología , Humanos
16.
J Vis Exp ; (61)2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22433991

RESUMEN

Human embryonic stem cells (hESC) are emerging as an attractive alternative source for cell replacement therapy since they can be expanded in culture indefinitely and differentiated to any cell types in the body. Various types of biomaterials have also been used in stem cell cultures to provide a microenvironment mimicking the stem cell niche(1-3). The latter is important for promoting cell-to-cell interaction, cell proliferation, and differentiation into specific lineages as well as tissue organization by providing a three-dimensional (3D) environment(4) such as encapsulation. The principle of cell encapsulation involves entrapment of living cells within the confines of semi-permeable membranes in 3D cultures(2). These membranes allow for the exchange of nutrients, oxygen and stimuli across the membranes, whereas antibodies and immune cells from the host that are larger than the capsule pore size are excluded(5). Here, we present an approach to culture and differentiate hESC DA neurons in a 3D microenvironment using alginate microcapsules. We have modified the culture conditions(2) to enhance the viability of encapsulated hESC. We have previously shown that the addition of p160-Rho-associated coiled-coil kinase (ROCK) inhibitor, Y-27632 and human fetal fibroblast-conditioned serum replacement medium (hFF-CM) to the 3D platform significantly enhanced the viability of encapsulated hESC in which the cells expressed definitive endoderm marker genes(1). We have now used this 3D platform for the propagation of hESC and efficient differentiation to DA neurons. Protein and gene expression analyses after the final stage of DA neuronal differentiation showed an increased expression of tyrosine hydroxylase (TH), a marker for DA neurons, >100 folds after 2 weeks. We hypothesized that our 3D platform using alginate microcapsules may be useful to study the proliferation and directed differentiation of hESC to various lineages. This 3D system also allows the separation of feeder cells from hESC during the process of differentiation and also has potential for immune-isolation during transplantation in the future.


Asunto(s)
Alginatos/química , Técnicas de Cultivo de Célula/métodos , Neuronas Dopaminérgicas/citología , Células Madre Embrionarias/citología , Andamios del Tejido , Cápsulas , Diferenciación Celular/fisiología , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos
17.
Stem Cells Dev ; 21(7): 997-1006, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22122714

RESUMEN

Alzheimer's disease (AD) is a leading cause of age-related dementia that is characterized by an extensive loss of neurons and synaptic transmission. The pathological hallmarks of AD are neurofibrillary tangles and deposition of ß-amyloid (Aß) plaques. Previous research has investigated how Aß fragments disrupt synaptic mechanisms in the vulnerable regions of the brain. There is a tremendous potential for stem cell technology to extend upon this research, not only in terms of developing therapeutic applications, but also in modeling AD. Indeed, the advent of induced pluripotent stem cell technology has opened up exciting new avenues for generating patient and disease-specific cell lines from somatic cells that may be used to model AD. Amyloid precursor protein (APP) is a key protein in neuronal development and this article reviews the role of APP in AD. Stem cell technology offers the opportunity to make use of APP in the directed differentiation of induced pluripotent stem cells into functional neurons, a process that may help generate a model of AD and thereby facilitate an understanding of the mechanisms underlying this disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Neurogénesis , Células Madre Pluripotentes/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Placa Amiloide/metabolismo , Células Madre Pluripotentes/metabolismo , Procesamiento Proteico-Postraduccional
18.
Pancreas ; 41(1): 54-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22143343

RESUMEN

OBJECTIVE: A challenge in using human embryonic stem cells (hESCs) as the source of surrogate ß cells is the establishment of methods that could effectively direct their differentiation into functional ß cells. The aim of this study was to assess the effect of NANOG gene suppression in differentiating hESCs as a mean of increasing the efficiency with which endoderm-derived pancreatic cells could be generated. METHODS: A homogenous cell population with stable suppression of NANOG was generated in hESC ENVY line using plasmid-based siRNA approach. Pancreatic differentiation was undertaken according to the ontology-based in vitro selection protocol and followed by transplantation into immunodeficiency mice to mature in vivo. RESULTS: We observed up-regulation of definitive endoderm genes, which expand the role of NANOG in blocking definitive endoderm differentiation. The ontology-based differentiation protocol resulted in increased expression of markers essential for pancreatic epithelium development. Transplantation of these cells further revealed a homogenous pancreatic exocrine-like morphology that stained positively for amylase. CONCLUSIONS: The suppression of NANOG displayed an effective differentiation toward endoderm and pancreatic progenitors. Investigation of the factors required for endocrine formation combined with a prolonged in vivo culturing could be further used to increase the ratio of endocrine-exocrine cells fate.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Endodermo/citología , Proteínas de Homeodominio/metabolismo , Páncreas/citología , Amilasas/metabolismo , Animales , Western Blotting , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía de Contraste de Fase , Proteína Homeótica Nanog , Páncreas/metabolismo , Páncreas Exocrino/citología , Páncreas Exocrino/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre/métodos , Células Madre/citología , Células Madre/metabolismo
19.
Stem Cells Dev ; 21(2): 206-16, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21506733

RESUMEN

Use of animal feeder layers and serum containing media in the derivation and propagation of induced pluripotent stem cells (iPSCs) can hinder clinical translation, because of the presence of xeno-material/pathogens. A defined and standardized system would be ideal for generating a homogenous population of iPSCs, which closely resembles human embryonic stem cells (hESCs). This article presents a novel and extensive comparison between in-house produced iPSCs and hESCs under "feeder" and "feeder-free" conditions, using transcriptomic genome-wide microarray analysis. We generated a list of pluripotency-associated and bivalent domain-containing genes by meta-analysis to measure qualitatively the degree of reprogramming in feeder-free derived iPSCs, in which both profiles displayed similar levels of gene expression as in hESCs. Gene ontology analysis showed that feeder-free iPSCs have enriched terms belonging to DNA repair/replication and cell cycle, which are signature to pluripotent cells. Transcriptomic data combined with directed differentiation assays, indicated that variability among iPSC lines is minimized when using a feeder-free cultural system, which may serve as a platform for further developing regenerative medicine compliant human iPSCs.


Asunto(s)
Técnicas de Cultivo de Célula , Reparación del ADN , Replicación del ADN , ADN/genética , Expresión Génica , Células Madre Pluripotentes Inducidas , Biomarcadores/análisis , Ciclo Celular , Diferenciación Celular , Línea Celular , Daño del ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Nutrientes , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcriptoma
20.
Regen Med ; 6(2): 215-27, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21391855

RESUMEN

Stem cells have been studied extensively for decades and they have the inherent capacity to self-renew as well as to generate one or more types of specialized cells. The current focus of research on stem cells, particularly on embryonic stem cells, is on directed differentiation of these cells into specific cell types for future regenerative medicine. For the past few years, the process of reprogramming, which mediates conversion of somatic cells to their pluripotent state, has been given much attention, as it provides a possible source of autologous stem cells. In addition, understanding the molecular mechanism of differentiation and reprogramming has long been a subject of interest. In this article, we have briefly introduced stem cells and discussed the use of embryonic stem cells in reprogramming of somatic cells and differentiation to different lineages. The application of embryonic stem cells extracts in inducing reprogramming and transdifferentiation has also been described and discussed. Should this approach be successful, patient-specific cells will be produced safely and the likelihood of rejection will be decreased when used in cell therapy for many debilitating human diseases for which there is no cure such as Parkinson's disease, Alzheimer's disease, diabetes and others.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico , Reprogramación Celular/efectos de los fármacos , Células Madre Embrionarias/química , Células Madre Embrionarias/citología , Animales , Bioingeniería/métodos , Bioingeniería/tendencias , Reprogramación Celular/fisiología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA