Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
1.
Cytotechnology ; 68(4): 1009-18, 2016 Aug.
Article de Anglais | MEDLINE | ID: mdl-25698160

RÉSUMÉ

Human skin has continuous self-renewal potential throughout adult life and serves as first line of defence. Its cellular components such as human epidermal keratinocytes (HEKs) and dermal mesenchymal stromal cells (DMSCs) are valuable resources for wound healing applications and cell based therapies. Here we show a simple, scalable and cost-effective method for sequential isolation and propagation of HEKs and DMSCs under defined culture conditions. Human skin biopsy samples obtained surgically were cut into fine pieces and cultured employing explant technique. Plated skin samples attached and showed outgrowth of HEKs. Gross microscopic examination displayed polygonal cells with a granular cytoplasm and H&E staining revealed archetypal HEK morphology. RT-PCR and immunocytochemistry authenticated the presence of key HEK markers including trans-membrane protein epithelial cadherin (E-cadherin), keratins and cytokeratin. After collection of HEKs by trypsin-EDTA treatment, mother explants were left intact and cultured further. Interestingly, we observed the appearance of another cell type with fibroblastic or stromal morphology which were able to grow up to 15 passages in vitro. Growth pattern, expression of cytoskeletal protein vimentin, surface proteins such as CD44, CD73, CD90, CD166 and mesodermal differentiation potential into osteocytes, adipocytes and chondrocytes confirmed their bonafide mesenchymal stem cell like status. These findings albeit preliminary may open up significant opportunities for novel applications in wound healing.

2.
Cytotherapy ; 18(1): 13-24, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26631828

RÉSUMÉ

The unique properties of mesenchymal stromal/stem cells (MSCs) to self-renew and their multipotentiality have rendered them attractive to researchers and clinicians. In addition to the differentiation potential, the broad repertoire of secreted trophic factors (cytokines) exhibiting diverse functions such as immunomodulation, anti-inflammatory activity, angiogenesis and anti-apoptotic, commonly referred to as the MSC secretome, has gained immense attention in the past few years. There is enough evidence to show that the one important pathway by which MSCs participate in tissue repair and regeneration is through its secretome. Concurrently, a large body of MSC research has focused on characterization of the MSC secretome; this includes both soluble factors and factors released in extracellular vesicles, for example, exosomes and microvesicles. This review provides an overview of our current understanding of the MSC secretome with respect to their potential clinical applications.


Sujet(s)
Cellules souches mésenchymateuses/métabolisme , Régénération , Mouvement cellulaire , Exosomes/métabolisme , Humains , Cellules souches mésenchymateuses/cytologie , Niche de cellules souches ,
3.
Exp Dermatol ; 24(5): 391-3, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25690925

RÉSUMÉ

Vitiligo is an autoimmune disorder that leads to depigmentation of skin via melanocyte dysfunction. Keratinocyte-induced toxicity is one among the several etiological factors implicated for vitiligo, and hence, autologous keratinocyte grafting is projected as one of the primary mode of treatment for vitiligo. However, reports indicate that perilesional keratinocytes not only display signatures of apoptosis but also could secrete cytokines and mediators which have antagonistic effect on proliferation or survival. Therefore, we investigated how vitiligo patients' derived keratinocytes respond to surplus amounts of inflammatory cytokines and whether they recapitulate events that take place during conventional wound healing. The primary objective of our study was to determine whether keratinocytes isolated from a vitiligo patient would undergo epithelial-mesenchymal transition similar to their normal counterparts upon induction with inflammatory cytokines such as TGF-b1 and EGF. We found that these keratinocytes undergo EMT during wound repair accompanied with increase in the levels of mesenchymal markers and ECM proteins; decrease in the levels of epithelial markers and enhanced migratory ability. Besides, we also demonstrated that EMT induction leads to activation of SMAD and MAPK pathways via Ras, Raf, PAI 1, Snail, Slug and ZO1. To our knowledge, this is the first report on the characterization of primary keratinocytes isolated from vitiligo patients with respect to their wound healing capacity.


Sujet(s)
Transition épithélio-mésenchymateuse , Kératinocytes/anatomopathologie , Vitiligo/anatomopathologie , Cicatrisation de plaie , Apoptose , Cellules cultivées , Cytokines/métabolisme , Transition épithélio-mésenchymateuse/physiologie , Humains , Kératinocytes/physiologie , Système de signalisation des MAP kinases , Modèles biologiques , Protéines Smad/métabolisme , Vitiligo/physiopathologie , Cicatrisation de plaie/physiologie
4.
Biotechnol Lett ; 37(1): 227-33, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25257585

RÉSUMÉ

To explore a novel source for the derivation of islets, we examined the differentiation potential of human non-pancreatic cancer cell lines, HeLa (cervical carcinoma cell line) and MCF-7 (breast cancer cell line). These cells were subjected to a serum-free, three-step sequential differentiation protocol which gave two distinct cell populations: single cells and cellular aggregates. Subsequent analysis confirmed their identity as pancreatic acinar cells and islet-like cell aggregates (ICAs), as evidenced by amylase secretion and diphenylthiocarbazone staining respectively. Reverse transcriptase-PCR and immunocytochemistry assessment of the ICAs revealed the expression of pancreatic specific markers Ngn-3, Glut-2, Pax-6 and Isl-1. These ICAs secreted insulin in response to glucose challenge, confirming their functionality. We propose that ICAs generated from HeLa and MCF-7 cell lines could form a promising in vitro platform of human islet equivalents (hIEQs) for diabetes research.


Sujet(s)
Différenciation cellulaire/physiologie , Ilots pancréatiques/cytologie , Ilots pancréatiques/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Amylases/métabolisme , Lignée cellulaire tumorale , Milieux de culture , Glucose/pharmacologie , Cellules HeLa , Humains , Ilots pancréatiques/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/effets des médicaments et des substances chimiques
5.
Med Hypotheses ; 83(6): 787-91, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25456787

RÉSUMÉ

Stem cell transplantation is a generic term covering different techniques. However there is argument over the pros and cons of autologous and allogeneic transplants of mesenchymal stem cells (MSCs) for regenerative therapy. Given that the MSCs have already been proven to be safe in patients, we hypothesize that allogeneic transplantation could be more effective and cost-effective as compared to autologous transplantation specifically in older subjects who are the likely victims of degenerative diseases. This analysis is based on the scientific logic that allogeneic stem cells extracted in large numbers from young and healthy donors could be physiologically, metabolically and genetically more stable. Therefore stem cells from young donors may be expected to exhibit higher vigor in secreting trophic factors leading to activation of host tissue-specific stem cells and also be more efficient in remodeling the micro-environmental niche of damaged tissue.


Sujet(s)
Médecine régénérative/méthodes , Transplantation de cellules souches/méthodes , Facteurs âges , Allogreffes , Cellules de la moelle osseuse/cytologie , Différenciation cellulaire , Vieillissement de la cellule , Analyse coût-bénéfice , Transplantation de cellules souches hématopoïétiques/économie , Transplantation de cellules souches hématopoïétiques/méthodes , Humains , Cellules souches mésenchymateuses/cytologie , Espèces réactives de l'oxygène/métabolisme , Médecine régénérative/économie , Transduction du signal , Transplantation de cellules souches/économie , Donneurs de tissus , Transplantation homologue
6.
J Biosci ; 39(1): 157-69, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24499800

RÉSUMÉ

Regenerative medicine is an evolving interdisciplinary topic of research involving numerous technological methods that utilize stem cells to repair damaged tissues. Particularly, mesenchymal stem cells (MSCs) are a great tool in regenerative medicine because of their lack of tumorogenicity, immunogenicity and ability to perform immunomodulatory as well as anti-inflammatory functions. Numerous studies have investigated the role of MSCs in tissue repair and modulation of allogeneic immune responses. MSCs derived from different sources hold unique regenerative potential as they are self-renewing and can differentiate into chondrocytes, osteoblasts, adipocytes, cardiomyocytes, hepatocytes, endothelial and neuronal cells, among which neuronal-like cells have gained special interest. MSCs also have the ability to secrete multiple bioactive molecules capable of stimulating recovery of injured cells and inhibiting inflammation. In this review we focus on neural differentiation potential of MSCs isolated from different sources and how certain growth factors/small molecules can be used to derive neuronal phenotypes from MSCs. We also discuss the efficacy of MSCs when transplanted in vivo and how they can generate certain neurons and lead to relief or recovery of the diseased condition. Furthermore, we have tried to evaluate the appropriatemerits of different sources ofMSCs with respect to their propensity towards neurological differentiation as well as their effectiveness in preclinical studies.


Sujet(s)
Différenciation cellulaire/physiologie , Protéines et peptides de signalisation intercellulaire/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/classification , Cellules souches mésenchymateuses/physiologie , Neurogenèse/physiologie , Médecine régénérative/méthodes , Humains , Modèles biologiques , Spécificité d'organe , Médecine régénérative/tendances
7.
Methods Mol Biol ; 1006: 147-66, 2013.
Article de Anglais | MEDLINE | ID: mdl-23546790

RÉSUMÉ

Several molecular methods like real-time PCR (Q-PCR), expression sequence tag (EST) scan, microarray and microRNA analysis, and massively parallel signature sequencing (MPSS) have proved to be increasingly sensitive and efficient for monitoring human embryonic stem cell (hESC) differentiation. However, most of these high-throughput tests have a limited use due to high cost, extended turnaround time, and the involvement of highly specialized technical expertise. Hence, there is a need of rapid, cost-effective, robust, yet sensitive method for routine screening of hESCs. A critical requirement in hESC cultures is to maintain a uniform undifferentiated state and to determine their differentiation capacity by showing the expression of germ-layer-specific gene markers. To determine the modulation of gene expression in hESCs during propagation, expansion, and differentiation via embryoid body (EB) formation, we developed a simple, rapid, inexpensive, and definitive multimarker, semiquantitative multiplex RT-PCR (mxPCR) platform technology. Among the 15 gene primers tested, 4 were pluripotent markers comprising of set 1; and 3 lineage-specific markers from each ecto-, meso-, and endoderm layers were combined as sets 2, 3, and 4, respectively. In summary, this study was performed to characterize hESCs on a molecular level and to determine the quality and degree of variability among hESC and their early progenies (EB). This single-reaction mxPCR assay was flexible and, by selecting appropriate reporter genes, can be designed for characterization of different hESC lines during routine maintenance and directed differentiation.


Sujet(s)
Marqueurs biologiques/métabolisme , Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Analyse de profil d'expression de gènes , Réaction de polymérisation en chaine multiplex/méthodes , Lignée cellulaire , Corps embryoïdes , Cellules souches embryonnaires/métabolisme , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes au cours du développement , Feuillets embryonnaires/métabolisme , Humains , Biologie moléculaire/méthodes
8.
J Biosci Bioeng ; 115(2): 200-6, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23040993

RÉSUMÉ

Post-myocardial infarction cardiomyocytes are the most important target cell types for cardiac repair. Many of the applications envisaged for human embryonic stem cells (hESC)-derived cardiomyocytes demand that the differentiation procedure be robust, cost effective and high yielding. Various lines of evidence including our earlier study suggest that hESCs have distinct preferences to become heart cells. However, a direct comparison between different protocols has not yet been reported to date. Here, we performed a logical and systematic comparison of cardiomyocytes obtained from hESCs via embryoid bodies (EBs) in suspension versus adherent static cultures of feeder-free hES colonies representing three-dimensional (3-D) and two-dimensional (2-D) culture systems, respectively. An in-depth characterization of the beating cells revealed appropriate cardiac marker expression both at gene and protein levels. Despite using similar media, 3-D and 2-D cultures showed significant variation in growth and ability to form beating areas. While the expression of pre-cardiac mesoderm markers like GATA-4, HAND1, Myf5, Msx1, and BMP-IIR remained unaltered; levels of functional heart-specific markers such as MLC-2A/2V, cTnT, ANP, Phospholamban, α-MHC and KV4.3 were substantially up-regulated in 3-D compared to 2-D cultures. Concurrently we observed a sharp decline in the expression of ESC, ectoderm and endoderm markers including Oct-4, Sox-2, NFH, Sox-1, Sox-17 and AFP. Further immunocytochemistry and flow cytometry demonstrated a higher percentage of cells positive for Brachyury, desmin and cardiac troponin in 3-D cultures. Our results underscore the higher efficiency of cardiomyocytes derived via 3-D cultures. This finding enriches our basic understanding of the differentiation pattern in hESC-derived cardiomyocytes.


Sujet(s)
Techniques de culture cellulaire/méthodes , Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Myocytes cardiaques/cytologie , Marqueurs biologiques/métabolisme , Lignée cellulaire , Ectoderme/métabolisme , Corps embryoïdes/cytologie , Cellules souches embryonnaires/métabolisme , Endoderme/métabolisme , Protéines foetales/métabolisme , Analyse de profil d'expression de gènes , Humains , Mésoderme/métabolisme , Myocytes cardiaques/métabolisme , Facteur de transcription Oct-3/métabolisme , Reproductibilité des résultats , Facteurs de transcription SOX/métabolisme , Protéines à domaine boîte-T/métabolisme , Facteurs temps
9.
CNS Neurosci Ther ; 19(1): 5-11, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-23253099

RÉSUMÉ

Neurodegenerative diseases are devastating because they cause increasing loss of cognitive and physical functions and affect an estimated 1 billion individuals worldwide. Unfortunately, no drugs are currently available to halt their progression, except a few that are largely inadequate. This mandates the search of new treatments for these progressively degenerative diseases. Neural stem cells (NSCs) have been successfully isolated, propagated, and characterized from the adult brains of mammals, including humans. The confirmation that neurogenesis occurs in the adult brain via NSCs opens up fresh avenues for treating neurological problems. The proof-of-concept studies demonstrating the neural differentiation capacity of stem cells both in vitro and in vivo have raised widespread enthusiasm toward cell-based interventions. It is anticipated that cell-based neurogenic drugs may reverse or compensate for deficits associated with neurological diseases. The increasing interest of the private sector in using human stem cells in therapeutics is evidenced by launching of several collaborative clinical research activities between Pharma giants and research institutions or small start-up companies. In this review, we discuss the major developments that have taken place in this field to position stem cells as a prospective candidate drug for the treatment of neurological disorders.


Sujet(s)
Transplantation cellulaire/méthodes , Transplantation cellulaire/tendances , Maladies du système nerveux/chirurgie , Animaux , Différenciation cellulaire/physiologie , Humains , Cellules souches neurales/physiologie , Cellules souches pluripotentes/physiologie
10.
J Transl Med ; 10: 229, 2012 Nov 21.
Article de Anglais | MEDLINE | ID: mdl-23171323

RÉSUMÉ

BACKGROUND: Numerous preclinical and clinical studies have investigated the regenerative potential and the trophic support of mesenchymal stem cells (MSCs) following their injection into a target organ. Clinicians favor the use of smallest bore needles possible for delivering MSCs into vascular organs like heart, liver and spleen. There has been a concern that small needle bore sizes may be detrimental to the health of these cells and reduce the survival and plasticity of MSCs. METHODS: In this report, we aimed to investigate the smallest possible bore size needle which would support the safe delivery of MSCs into various tissues for different clinical or cosmetic applications. To accomplish this we injected cells via needle sizes 24, 25 and 26 G attached to 1 ml syringe in the laboratory and collected the cells aseptically. Control cells were ejected via 1 ml syringe without any needle. Thereafter, the needle ejected cells were cultured and characterized for their morphology, attachment, viability, phenotypic expression, differentiation potential, cryopreservation and in vivo migration abilities. In the second phase of the study, cells were injected via 26 G needle attached to 1 ml syringe for 10 times. RESULTS: Similar phenotypic and functional characteristics were observed between ejected and control group of cells. MSCs maintained their cellular and functional properties after single and multiple injections. CONCLUSIONS: This study proves that 26 G bore size needles can be safely used to inject MSCs for clinical/therapeutics purposes.


Sujet(s)
Cosmétiques , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/cytologie , Aiguilles , Adulte , Animaux , Marqueurs biologiques/métabolisme , Cellules de la moelle osseuse/cytologie , Carbocyanines/métabolisme , Adhérence cellulaire , Différenciation cellulaire , Membrane cellulaire/métabolisme , Mouvement cellulaire , Forme de la cellule , Survie cellulaire , Vieillissement de la cellule , Cryoconservation , Humains , Injections , Phénotype , Rats , Rat nude , Coloration et marquage , Jeune adulte
11.
Cytotherapy ; 14(8): 902-16, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22731756

RÉSUMÉ

Critical limb ischemia (CLI) is a syndrome manifested by ischemic rest pain, non-healing ulcers and tissue loss. CLI patients are at very high risk of amputation and experience poor physical function, leading to severe morbidity and mortality. The fundamental goal for CLI treatment is to relieve ischemic rest pain, heal ulcers, prevent limb loss and improve the quality of life, thereby extending the survival of the patient. Surgical or endovascular revascularization aimed at increasing blood flow is currently available for limb salvage in CLI. However, up to 30% of CLI patients are not suitable for such interventions because of high operative risk or unfavorable vascular anatomy. Therefore exploring new and more effective strategies for revascularization of ischemic limbs is imperative for the establishment of a viable therapeutic alternative. With the emergence of new approaches, this review describes up-to-date progress and developments in cell-based therapy as a novel and promising alternative for CLI treatment. Preliminary clinical data have established the safety, feasibility and efficacy of stem cells, and numerous studies are underway to consolidate this evidence further. However, significant hurdles remain to be addressed before this research can be responsibly translated to the bedside. In particular, we need better understanding of the behavior of cells post-transplantation and to learn how to control their survival and migration proliferation/differentiation in the hostile pathologic environment. Future research should focus on methods of isolation, optimal dosage, appropriate cell type, route of administration, role of tissue-derived factors and supportive endogenous stimulation.


Sujet(s)
Thérapie cellulaire et tissulaire , Membres/physiopathologie , Ischémie/thérapie , Cellules souches , Différenciation cellulaire , Prolifération cellulaire , Membres/vascularisation , Humains , Ischémie/physiopathologie , Néovascularisation physiologique , Transplantation de cellules souches , Cellules souches/classification , Cellules souches/cytologie
12.
J Cell Biochem ; 113(10): 3153-64, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22615164

RÉSUMÉ

The clinical application of human bone marrow derived multipotent mesenchymal stromal cells (MSC) requires expansion, cryopreservation, and transportation from the laboratory to the site of cell implantation. The cryopreservation and thawing process of MSCs may have important effects on the viability, growth characteristics and functionality of these cells both in vitro and in vivo. More importantly, MSCs after two rounds of cryopreservation have not been as well characterized as fresh MSCs from the transplantation perspective. The objective of this study was to determine if the effect of successive cryopreservation of pooled MSCs during the exponential growth phase could impair their morphology, phenotype, gene expression, and differentiation capabilities. MSCs cryopreserved at passage 3 (cell bank) were thawed and expanded up to passage 4 and cryopreserved for the second time. These cells (passive) were then thawed and cultured up to passage 6, and, at each passage MSCs were characterized. As control, pooled passage 3 cells (active) after one round of cryopreservation were taken all the way to passage 6 without cryopreservation. We determined the growth rate of MSCs for both culture conditions in terms of population doubling number (PDN) and population doubling time (PDT). Gene expression profiles for pluripotency markers and tissue specific markers corresponding to neuroectoderm, mesoderm and endoderm lineages were also analyzed for active and passive cultures of MSC. The results show that in both culture conditions, MSCs exhibited similar growth properties, phenotypes and gene expression patterns as well as similar differentiation potential to osteo-, chondro-, and adipo-lineages in vitro. To conclude, it appears that successive or multiple rounds of cryopreservation of MSCs did not alter the fundamental characteristics of these cells and may be used for clinical therapy.


Sujet(s)
Moelle osseuse/métabolisme , Cryoconservation/méthodes , Cellules souches mésenchymateuses/cytologie , Marqueurs biologiques , Moelle osseuse/physiologie , Numération cellulaire , Techniques de culture cellulaire/méthodes , Différenciation cellulaire , Lignage cellulaire , Prolifération cellulaire , Survie cellulaire , Vieillissement de la cellule , Milieux de culture/métabolisme , Humains , Immunophénotypage , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/physiologie , Phénotype , Température , Facteurs temps , Transcriptome
13.
Arch Toxicol ; 86(4): 651-61, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22105179

RÉSUMÉ

In vitro disease modeling using pluripotent stem cells can be a fast track screening tool for toxicological testing of candidate drug molecules. Dimethyl sulfoxide (DMSO) is one of the most commonly used solvents in drug screening. In the present investigation, we exposed 14- to 21-day-old embryoid bodies (EBs) to three different concentrations of DMSO [0.01% (low dose), 0.1% (medium dose) and 1.0% (high dose)] to identify the safest dose that could effectively be used as solvent. We found that DMSO treatment substantially altered the morphology and attachment of cells in concurrence with a significant reduction in cell viability in a dose-dependent manner. Gene expression studies revealed a selective downregulation of key markers associated with stemness (Oct-4, Sox-2, Nanog and Rex-1); ectoderm (Nestin, TuJ1, NEFH and Keratin-15); mesoderm (HAND-1, MEF-2C, GATA-4 and cardiac-actin); and endoderm (SOX-17, HNF-3ß, GATA-6 and albumin), indicating an aberrant and untimely differentiation trajectory. Furthermore, immunocytochemistry, flow cytometry and histological analyses demonstrated substantial decrease in the levels of albumin and CK-18 proteins coupled with a massive reduction in the number of cells positive for PAS staining, implicating reduced deposits of glycogen. Our study advocates for the first time that DMSO exposure not only affects the phenotypic characteristics but also induces significant alteration in gene expression, protein content and functionality of the differentiated hepatic cells. Overall, our experiments warrant that hESC-based assays can provide timely alerts about the outcome of widespread applications of DMSO as drug solvent, cryoprotectant and differentiating agent.


Sujet(s)
Cryoprotecteurs/toxicité , Diméthylsulfoxyde/toxicité , Corps embryoïdes/effets des médicaments et des substances chimiques , Solvants/toxicité , Marqueurs biologiques/métabolisme , Adhérence cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Corps embryoïdes/métabolisme , Corps embryoïdes/anatomopathologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glycogène/métabolisme , Humains
14.
J Cell Biochem ; 113(1): 19-30, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-21956183

RÉSUMÉ

The in vitro derived hepatocytes from human embryonic stem cells (hESC) is a promising tool to acquire improved knowledge of the cellular and molecular events underlying early human liver development under physiological and pathological conditions. Here we report a simple two-step protocol employing conditioned medium (CM) from human hepatocellular carcinoma cell line, HepG2 to generate functional hepatocyte-like cells from hESC. Immunocytochemistry, flow cytometry, quantitative RT-PCR, and biochemical analyses revealed that the endodermal progenitors appeared as pockets in culture, and the cascade of genes associated with the formation of definitive endoderm (HNF-3ß, SOX-17, DLX-5, CXCR4) was consistent and in concurrence with the up-regulation of the markers for hepatic progenitors [alpha-feto protein (AFP), HNF-4α, CK-19, albumin, alpha-1-antitrypsin (AAT)], followed by maturation into functional hepatocytes [tyrosine transferase (TAT), tryptophan-2, 3-dioxygenase (TDO), glucose 6-phosphate (G6P), CYP3A4, CYP7A1]. We witnessed that the gene expression profile during this differentiation process recapitulated in vivo liver development demonstrating a gradual down-regulation of extra embryonic endodermal markers (SOX-7, HNF-1ß, SNAIL-1, LAMININ-1, CDX2), and the generated hepatic cells performed multiple liver functions. Since prenatal alcohol exposure is known to provoke irreversible abnormalities in the fetal cells and developing tissues, we exposed in vitro generated hepatocytes to ethanol (EtOH) and found that EtOH treatment not only impairs the survival and proliferation, but also induces apoptosis and perturbs differentiation of progenitor cells into hepatocytes. This disruption was accompanied by alterations in the expression of genes and proteins involved in hepatogenesis. Our results provide new insights into the wider range of destruction caused by alcohol on the dynamic process of liver organogenesis.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules souches embryonnaires/cytologie , Hépatocytes/cytologie , Hépatocytes/physiologie , Marqueurs biologiques , Lignée cellulaire , Survie cellulaire , Milieux de culture conditionnés/pharmacologie , Éthanol/métabolisme , Analyse de profil d'expression de gènes , Cellules HepG2 , Hépatocytes/métabolisme , Humains , Foie/embryologie , Foie/métabolisme , Maladies alcooliques du foie/anatomopathologie , Organogenèse/physiologie
15.
CNS Neurol Disord Drug Targets ; 10(6): 741-56, 2011 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-21838668

RÉSUMÉ

Stroke causes a devastating insult to the brain resulting in severe neurological deficits because of a massive loss of different neurons and glia. In the United States, stroke is the third leading cause of death. Stroke remains a significant clinical unmet condition, with only 3% of the ischemic patient population benefiting from current treatment modalities, such as the use of thrombolytic agents, which are often limited by a narrow therapeutic time window. However, regeneration of the brain after ischemic damage is still active days and even weeks after stroke occurs, which might provide a second window for treatment. Neurorestorative processes like neurogenesis, angiogenesis and synaptic plasticity lead to functional improvement after stroke. Stem cells derived from various tissues have the potential to perform all of the aforementioned processes, thus facilitating functional recovery. Indeed, transplantation of stem cells or their derivatives in animal models of cerebral ischemia can improve function by replacing the lost neurons and glial cells and by mediating remyelination, and modulation of inflammation as confirmed by various studies worldwide. While initially stem cells seemed to work by a 'cell replacement' mechanism, recent research suggests that cell therapy works mostly by providing trophic support to the injured tissue and brain, fostering both neurogenesis and angiogenesis. Moreover, ongoing human trials have encouraged hopes for this new method of restorative therapy after stroke. This review describes up-to-date progress in cell-based therapy for the treatment of stroke. Further, as we discuss here, significant hurdles remain to be addressed before these findings can be responsibly translated to novel therapies. In particular, we need a better understanding of the mechanisms of action of stem cells after transplantation, the therapeutic time window for cell transplantation, the optimal route of cell delivery to the ischemic brain, the most suitable cell types and sources and learn how to control stem cell proliferation, survival, migration, and differentiation in the pathological environment. An integrated approach of cell-based therapy with early-phase clinical trials and continued preclinical work with focus on mechanisms of action is needed.


Sujet(s)
Transplantation de cellules souches/méthodes , Transplantation de cellules souches/tendances , Accident vasculaire cérébral/anatomopathologie , Accident vasculaire cérébral/thérapie , Animaux , Encéphalopathie ischémique/anatomopathologie , Encéphalopathie ischémique/thérapie , Humains , Dégénérescence nerveuse/anatomopathologie , Dégénérescence nerveuse/thérapie
16.
J Cell Biochem ; 112(5): 1353-63, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21337383

RÉSUMÉ

Among the different parameters governing the successful derivation and expansion of human embryonic stem cells (hESC), feeder layers play the most important role. Human feeders in form of human mesenchymal stromal cells (hMSCs) and human foreskin fibroblasts (HFFs) lay the foundation for eradication of animal-derived hESC culture system. In this study we explored the potential of human foreskin derived mesenchymal like stromal cells (HF-MSCs) to support self renewal and pluripotency of hESC. The MSCs isolated from human foreskin were found to be resistant to standard concentrations and duration of mitomycin-C treatment. Growth pattern, gene profiling (Oct-4, Nanog, Sox-2, Rex-1), cytoskeletal protein expression (vimentin, nestin) and tri-lineage differentiation potential into adipocytes, chondrocytes and osteocytes confirmed their mesenchymal stromal cell status. Further, the HF-MSCs were positive for CD105, CD166, CD73, CD44, CD90, SSEA-4, and negative for CD34, CD45, HLA-DR cell-surface markers and were found to exhibit BM-MSC-like characteristics. hESC lines co-cultured with HF-MSC feeders showed expression of expected pluripotent transcription factors Oct-4, Nanog, Sox-2, GDF-3, Rex-1, STELLAR, ABCG2, Dppa5, hTERT; surface markers SSEA-4, TRA-1-81 and maintained their cytogenetic stability during long term passaging. These novel feeders also improved the formation of embryoid bodies (EBs) from hESC which produced cell types representing three germ layers. This culture system has the potential to aid the development of clinical-grade hESCs for regenerative medicine and drug screening. Further, we envisage foreskin can serve as a valuable source of alternative MSCs for specific therapeutic applications.


Sujet(s)
Techniques de coculture , Cellules souches embryonnaires/cytologie , Mésoderme/cytologie , Cellules souches pluripotentes/cytologie , Adipogenèse/génétique , Adolescent , Techniques de culture cellulaire , Différenciation cellulaire , Prolifération cellulaire , Enfant , Cellules souches embryonnaires/métabolisme , Fibroblastes/cytologie , Fibroblastes/métabolisme , Prépuce/cytologie , Humains , Mâle , Mésoderme/métabolisme , Cellules souches pluripotentes/métabolisme , Cellules stromales/cytologie , Cellules stromales/métabolisme , Jeune adulte
17.
Med Hypotheses ; 76(4): 599-601, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21277690

RÉSUMÉ

The staggering number of publications featuring the use of stem cells has revolutionized regenerative medicine research. Preclinical studies indicate that allogeneic human mesenchymal stem cells (MSCs) may be useful for the treatment of several clinical disorders, including sepsis, acute renal failure, acute myocardial infarction, and more recently, acute lung injury (ALI). However, considerable success would not be obtained in clinical trials due to poor survival of transplanted cells under the influence of inflammatory conditions. Despite robust approaches like cellular reprogramming, scaffolds and conditioned media have been tested to overcome this problem; however the success rate of these approaches remain questionable. Recently, pretreatment of bioactive compounds in vitro have been shown to suppress cell apoptosis and promote cell survival. Quite likely a similar phenomenon can take place in vivo. Based on such studies, we hypothesize that MSCs derived from human post-natal tissues could be conditioned and prepared for targeted disease therapy. Depending on the disease condition, the MSCs could be treated prior to delivery with appropriate bioactive compounds to allow them survive longer and perform a better role as biocatalyst. The advantage of this approach could be the tailor made availability of MSCs preconditioned with appropriate bioactive compounds for disease specific therapy. Therefore, the choice of suitable bioactive molecule is likely to enhance the efficacy of targeted stem cell therapy and preconditioning may provide a novel strategy in maximizing biological and functional properties of MSCs.


Sujet(s)
Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/physiologie , Régénération/effets des médicaments et des substances chimiques , Atorvastatine , Survie cellulaire/effets des médicaments et des substances chimiques , Diazoxide/pharmacologie , Acides heptanoïques/pharmacologie , Humains , Pyrroles/pharmacologie , Médecine régénérative , Transplants , Trimétazidine/pharmacologie
18.
J Cell Physiol ; 226(6): 1583-95, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-20945368

RÉSUMÉ

In vitro models based on embryonic stem cells (ESC) are highly promising for improvement of predictive toxicology screening in humans. After the successful validation of embryonic stem cell test (EST) in 2001; concerns have been raised on the usage of mouse ESC and also the morphological evaluation of beating cell clusters. This requires specialized skill-sets and is highly prone to misjudgement and false positive results. To overcome these limitations, we undertook the present study incorporating improvisations over the conventional EST. Here, we explored the potential of a human ESC (hESC)-based assay to evaluate the potential toxicity of penicillin-G, caffeine, and hydroxyurea. Drug treatment inhibited hESC adhesion and substantially altered the morphology and viability (∼ 50%) of embryoid bodies (EBs). Flow cytometry analysis not only showed a significant increase of apoptotic cells in the highest doses but also induced a diverse pattern in DNA content and cell cycle distribution relative to control. Both semi-quantitative and quantitative RT-PCR studies revealed a selective down regulation of markers associated with stemness (Nanog, Rex1, SOX-2, and hTERT); cardiac mesoderm (Cripto1, MEF-2C, and Brachyury); hepatic endoderm (AFP, HNF-3ß, HNF-4α, GATA-4, and SOX-17); and neuroectoderm (Nestin, SOX-1, NURR1, NEFH, Synaptophysin, TH, and Olig2) in a drug as well as dose dependent manner indicating abnormal differentiation. Furthermore, a decrease in the expression of AFP and GFAP proteins followed by a dose-dependent reduction in the levels of hCG-ß, progesterone-II, and estradiol hormones was demonstrated by immunocytochemistry and ECLIA, respectively. This new and unique approach comprising of DNA cell cycle analysis, germ layer-specific marker expression and hormone levels as endpoints might offer a clinically relevant and commercially viable alternative for predicting in vivo developmental toxicity.


Sujet(s)
Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Tests de toxicité , Animaux , Marqueurs biologiques/métabolisme , Caféine , Adhérence cellulaire/effets des médicaments et des substances chimiques , Cycle cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Lignée cellulaire , Lignage cellulaire/effets des médicaments et des substances chimiques , Lignage cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Forme de la cellule/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Corps embryoïdes/cytologie , Corps embryoïdes/effets des médicaments et des substances chimiques , Corps embryoïdes/métabolisme , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Hormones/métabolisme , Humains , Hydroxy-urée/toxicité , Souris , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Spécificité d'organe/effets des médicaments et des substances chimiques , Spécificité d'organe/génétique , Benzylpénicilline/toxicité , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/métabolisme , Contrôle de qualité
19.
J Biomol Screen ; 15(6): 630-43, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20530724

RÉSUMÉ

Techniques to evaluate gene expression profiling, including real-time quantitative PCR, TaqMan low-density arrays, and sufficiently sensitive cDNA microarrays, are efficient methods for monitoring human embryonic stem cell (hESC) cultures. However, most of these high-throughput tests have a limited use due to high cost, extended turnaround time, and the involvement of highly specialized technical expertise. Hence, there is a paucity of rapid, cost-effective, robust, yet sensitive methods for routine screening of hESCs. A critical requirement in hESC cultures is to maintain a uniform undifferentiated state and to determine their differentiation capacity by showing the expression of gene markers representing all germ layers, including ecto-, meso-, and endoderm. To quantify the modulation of gene expression in hESCs during their propagation, expansion, and differentiation via embryoid body (EB) formation, the authors developed a simple, rapid, inexpensive, and definitive multimarker, semiquantitative multiplex RT-PCR (mxPCR) platform technology. Among the 15 gene primers tested, 4 were pluripotent markers comprising set 1, and 3 lineage-specific markers from each ecto-, meso-, and endoderm layers were combined as sets 2 to 4, respectively. The authors found that these 4 sets were not only effective in determining the relative differentiation in hESCs, but were easily reproducible. In this study, they used the HUES-7 cell line to standardize the technique, which was subsequently validated with HUES-9, NTERA-2, and mouse embryonic fibroblast cells. This single-reaction mxPCR assay was flexible and, by selecting appropriate reporter genes, can be designed for characterization of different hESC lines during routine maintenance and directed differentiation.


Sujet(s)
Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/métabolisme , Réaction de polymérisation en chaîne/méthodes , Animaux , Marqueurs biologiques/métabolisme , Différenciation cellulaire/génétique , Lignée cellulaire , Embryon de mammifère/cytologie , Embryon de mammifère/métabolisme , Technique d'immunofluorescence , Régulation de l'expression des gènes au cours du développement , Humains , Souris , Normes de référence , Reproductibilité des résultats
20.
Exp Biol Med (Maywood) ; 234(10): 1230-43, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19546356

RÉSUMÉ

Human embryonic stem cells (hESCs) are able to stably maintain their characteristics for an unlimited period; nevertheless, substantial differences among cell lines in gene and protein expression not manifested during the undifferentiated state may appear when cells differentiate. It is widely accepted that developing an efficient protocol to control the differentiation of hESCs will enable us to produce adequate numbers of desired cell types with relative ease for diverse applications ranging from basic research to cell therapy and drug screening. Hence of late, there has been considerable interest in understanding whether and how hESC lines are equivalent or different to each other in their in vitro developmental tendencies. In this study, we compared the developmental competences of two hESC lines (HUES-9 and HUES-7) at molecular, cellular and functional levels, upon spontaneous differentiation without any added inducing agents. Both cell lines generated the three embryonic germ layers, extra-embryonic tissues and primordial germ cells during embryoid body (EB) formation. However HUES-9 showed a stronger propensity towards formation of neuroectodermal lineages, whereas HUES-7 differentiated preferentially into mesoderm and endoderm. Upon further differentiation, HUES-9 generated largely neural cells (neurons, oligodendrocytes, astrocytes and gangliosides) whereas HUES-7 formed mesendodermal derivatives, including cardiomyocytes, skeletal myocytes, endothelial cells, hepatocytes and pancreatic cells. Overall, our findings endorse the hypothesis that independently-derived hESCs biologically differ among themselves, thereby displaying varying differentiation propensity. These subtle differences not only highlight the importance of screening and deriving lines for lineage-specific differentiation but also indicate that individual lines may possess a repertoire of capabilities that is unique.


Sujet(s)
Lignée cellulaire , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/physiologie , Marqueurs biologiques/métabolisme , Différenciation cellulaire , Lignage cellulaire , Cellules cultivées , Ectoderme/cytologie , Cellules souches embryonnaires/métabolisme , Endoderme/cytologie , Technique d'immunofluorescence indirecte , Humains , Immunohistochimie , Caryotypage , Mésoderme/cytologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...