Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
iScience ; 26(11): 108100, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37915594

ABSTRACT

Liver transplantation is the gold-standard therapy for acute hepatic failure (AHF) with limitations related to organ shortage and life-long immunosuppressive therapy. Cell therapy emerges as a promising alternative to transplantation. We have previously shown that IL-10 and Annexin-A1 released by amniotic fluid human mesenchymal stromal cells (AF-MSCs) and their hepatocyte progenitor-like (HPL) or hepatocyte-like (HPL) cells induce liver repair and downregulate systemic inflammation in a CCl4-AHF mouse model. Herein, we demonstrate that exosomes (EXO) derived from these cells improve liver phenotype in CCl4-induced mice and promote oval cell proliferation. LC-MS/MS proteomic analysis identified MEFG-8 in EXO cargo that facilitates rescue of AHF by suppressing PI3K signaling. Administration of recombinant MFGE-8 protein also reduced liver damage in CCl4-induced mice. Clinically, MEFG-8 expression was decreased in liver biopsies from AHF patients. Collectively, our study provides proof-of-concept for an innovative, cell-free, less immunogenic, and non-toxic alternative strategy for AHF.

2.
EBioMedicine ; 45: 542-552, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31303498

ABSTRACT

BACKGROUND: Human mesenchymal stem/stromal cells (MSCs) and their secreted molecules exert beneficial effects in injured tissues by promoting tissue regeneration and angiogenesis and by inhibiting inflammation and fibrosis. We have previously demonstrated that the therapeutic activity of fetal MSCs derived from amniotic fluid (AF-MSCs) and their hepatic progenitor-like cells (HPL) is mediated by paracrine effects in a mouse model of acute hepatic failure (AHF). METHODS: Herein, we have combined proteomic profiling of the AF-MSCs and HPL cell secretome with ex vivo and in vivo functional studies to identify specific soluble factors, which underpin tissue regeneration in AHF. FINDINGS: The anti-inflammatory molecule Annexin-A1 (ANXA1) was detected at high levels in both AF-MSC and HPL cell secretome. Further functional analyses revealed that the shRNA-mediated knock-down of ANXA1 in MSCs (shANXA1-MSCs) decreased their proliferative, clonogenic and migratory potential, as well as their ability to differentiate into HPL cells. Liver progenitors (oval cells) from AHF mice displayed reduced proliferation when cultured ex vivo in the presence of conditioned media from shANXA1-MSCs compared to control MSCs secretome. Intra-hepatic delivery of conditioned media from control MSCs but not shANXA1-MSCs reduced liver damage and circulating levels of pro-inflammatory cytokines in AHF. INTERPRETATION: Collectively, our study uncovers secreted Annexin-A1 as a novel effector of MSCs in liver regeneration and further underscores the potential of cell-free therapeutic strategies for liver diseases. FUND: Fondation Santé, GILEAD Asklipeios Grant, Fellowships of Excellence - Siemens, IKY, Reinforcement of Postdoctoral Researchers, IKY.


Subject(s)
Annexin A1/genetics , Liver Regeneration/genetics , Mesenchymal Stem Cell Transplantation , Proteomics , Animals , Annexin A1/metabolism , Bone Marrow Cells/metabolism , Cell Proliferation/genetics , Culture Media, Conditioned/pharmacology , Fetus , Humans , Intercellular Signaling Peptides and Proteins , Liver/metabolism , Liver/pathology , Mesenchymal Stem Cells/metabolism , Mice
3.
J Vis Exp ; (124)2017 06 09.
Article in English | MEDLINE | ID: mdl-28654077

ABSTRACT

Human pluripotent stem cells can differentiate into various cell types that can be applied to human-based in vitro toxicity assays. One major advantage is that the reprogramming of somatic cells to produce human induced pluripotent stem cells (hiPSCs) avoids the ethical and legislative issues related to the use of human embryonic stem cells (hESCs). HiPSCs can be expanded and efficiently differentiated into different types of neuronal and glial cells, serving as test systems for toxicity testing and, in particular, for the assessment of different pathways involved in neurotoxicity. This work describes a protocol for the differentiation of hiPSCs into mixed cultures of neuronal and glial cells. The signaling pathways that are regulated and/or activated by neuronal differentiation are defined. This information is critical to the application of the cell model to the new toxicity testing paradigm, in which chemicals are assessed based on their ability to perturb biological pathways. As a proof of concept, rotenone, an inhibitor of mitochondrial respiratory complex I, was used to assess the activation of the Nrf2 signaling pathway, a key regulator of the antioxidant-response-element-(ARE)-driven cellular defense mechanism against oxidative stress.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Cell Differentiation/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Rotenone/toxicity , Signal Transduction/drug effects , Toxicity Tests/methods
4.
Neurochem Int ; 108: 457-471, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28627367

ABSTRACT

Activation of Nrf2/ARE signaling pathway occurs ubiquitously in most cell types upon induction of oxidative stress. Rotenone, an inhibitor of mitochondrial complex I, can be used to trigger oxidative stress, stimulate the activation of Nrf2 pathway in neuronal and astrocytic cells and assess neurotoxicity. We have previously demonstrated that an acute treatment with rotenone can induce Nrf2 activation, which leads to astrocyte activation and dopaminergic (DA) neuronal cell death in a mixed neuronal/astrocytic cell model derived from human induced pluripotent stem cells (hiPSCs). In this study, we characterized the effects of a repeated dose treatment with rotenone (14 days) on hiPSC-derived neural stem cells (NSCs) undergoing differentiation, assessing the expression and the activation of the Nrf2 pathway. Our results show that Nrf2 signaling increases during NSC differentiation. Moreover, we observed that rotenone treatment induced a progressive activation of Nrf2 signaling together with a induction of astrocyte reactivity, a reduction of neurite length leading to neuronal cell death, in particular of DA neurons. Altogether these data indicate that hiPSC-NSC models are relevant test systems for the evaluation of Nrf2 pathway activation upon induced oxidative stress, allowing further understanding of the molecular mechanisms underlying exposure to (developmental) neurotoxicants.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Induced Pluripotent Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Neural Stem Cells/metabolism , Rotenone/toxicity , Astrocytes/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/metabolism , Uncoupling Agents/toxicity
5.
Stem Cells Dev ; 26(7): 482-494, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28068868

ABSTRACT

Recent findings indicate that microRNAs (miRNAs) are critical for the regulatory network of adipogenesis in human mesenchymal stem/stromal cells (MSCs). Fetal MSCs derived from amniotic fluid (AF-MSCs) represent a population of multipotent stem cells characterized by a wide range of differentiation properties that can be applied in cell-based therapies. In this study, miRNA microarray analysis was performed to assess miRNA expression in terminal differentiated AF-MSCs into adipocyte-like cells (AL cells). MiR-26a was identified in high expression levels in AL cells indicating a critical role in the process of adipogenesis. Overexpression of miR-26a in AF-MSCs led to significant induction of their adipogenic differentiation properties that were altered after miR-26a inhibition. We have demonstrated that miR-26a regulates adipogenesis through direct inhibition of PTEN, which in turn promotes activation of Akt pathway. Also, miR-26a modulates cell cycle during adipogenesis by interacting with Cyclin E1 and CDK6. These results point to the regulatory role of miR-26a and its target genes PTEN, Cyclin E1, and CDK6 in adipogenic differentiation of AF-MSCs, providing a basis for understanding the mechanisms of fat cell development and obesity.


Subject(s)
Adipogenesis/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 6/metabolism , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Multipotent Stem Cells/cytology , Oncogene Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Adipocytes/cytology , Adipose Tissue/cytology , Amniotic Fluid/metabolism , Cells, Cultured , Humans , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics
6.
Neurochem Int ; 106: 62-73, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27615060

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are considered as a powerful tool for drug and chemical screening and development of new in vitro testing strategies in the field of toxicology, including neurotoxicity evaluation. These cells are able to expand and efficiently differentiate into different types of neuronal and glial cells as well as peripheral neurons. These human cells-based neuronal models serve as test systems for mechanistic studies on different pathways involved in neurotoxicity. One of the well-known mechanisms that are activated by chemically-induced oxidative stress is the Nrf2 signaling pathway. Therefore, in the current study, we evaluated whether Nrf2 signaling machinery is expressed in human induced pluripotent stem cells (hiPSCs)-derived mixed neuronal/glial culture and if so whether it becomes activated by rotenone-induced oxidative stress mediated by complex I inhibition of mitochondrial respiration. Rotenone was found to induce the activation of Nrf2 signaling particularly at the highest tested concentration (100 nM), as shown by Nrf2 nuclear translocation and the up-regulation of the Nrf2-downstream antioxidant enzymes, NQO1 and SRXN1. Interestingly, exposure to rotenone also increased the number of astroglial cells in which Nrf2 activation may play an important role in neuroprotection. Moreover, rotenone caused cell death of dopaminergic neurons since a decreased percentage of tyrosine hydroxylase (TH+) cells was observed. The obtained results suggest that hiPSC-derived mixed neuronal/glial culture could be a valuable in vitro human model for the establishment of neuronal specific assays in order to link Nrf2 pathway activation (biomarker of oxidative stress) with additional neuronal specific readouts that could be applied to in vitro neurotoxicity evaluation.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Rotenone/toxicity , Signal Transduction/physiology , Animals , Cells, Cultured , Coculture Techniques , Drug Evaluation, Preclinical/methods , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Insecticides/toxicity , Neurons/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Signal Transduction/drug effects
7.
Stem Cells Dev ; 24(20): 2337-52, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26176180

ABSTRACT

In recent years it has been hypothesized that cancer stem cells (CSCs) are the actual driving force of tumor formation, highlighting the need to specifically target CSCs to successfully eradicate cancer growth and recurrence. Particularly, the deregulation of physiological signaling pathways controlling stem cell proliferation, self-renewal, differentiation, and metabolism is currently considered as one of the leading determinants of cancer formation. Given their peculiar, slow-dividing phenotype and their ability to respond to multiple microenvironmental stimuli, stem cells appear to be more susceptible to genetic and epigenetic carcinogens, possibly undergoing mutations resulting in tumor formation. In particular, some animal-derived bioactive nutrients and metabolites known to affect the hormonal milieu, and also chemicals derived from food processing and cooking, have been described as possible carcinogenic factors. Here, we review most recent literature in this field, highlighting how some environmental toxicants, some specific nutrients and their secondary products can induce carcinogenesis, possibly impacting stem cells and their niches, thus causing tumor growth.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens, Environmental/adverse effects , Cell Differentiation/drug effects , Food/adverse effects , Neoplastic Stem Cells/cytology , Animals , Carcinogenesis/metabolism , Cell Differentiation/physiology , Humans , Neoplasm Recurrence, Local/pathology
8.
Toxicol Lett ; 231(1): 38-44, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25192806

ABSTRACT

A number of in vitro toxicity assays based on human embryonic stem cells (hESCs) are under development in order to provide alternative methods for the screening of chemicals and drugs and to reduce the number of animals needed for developmental toxicity assessment. The major challenge is to demonstrate the reliability of these in vitro methods by correlating the in vitro produced results to the available in vivo data. In this context transcriptomic approaches associated to toxicogenomic database analysis give the possibility to screen, annotate and cluster high numbers of genes and to identify the molecular changes that univocally mark the toxicity induced processes or are indicative of the early initiating events that lead to cellular toxicity. In this retrospective study we compare microarray transcriptomic data derived from two different hESCs lines (HUES1 and H9) exposed to valproic acid (VA) while applying the same differentiation protocol. We present the results of this comparative analysis in light of the known teratogenic effects of VA. The results show molecular changes in the processes of neural development, neural crest migration, apoptosis and regulation of transcription, indicating a good correspondence with the available in vivo data. We also describe common toxicological signatures and provide an interpretation of the observed qualitative differences referring to known biological features of the two hESCs lines.


Subject(s)
Embryonic Stem Cells/drug effects , Gene Expression Profiling/methods , Neural Stem Cells/drug effects , Oligonucleotide Array Sequence Analysis , Toxicity Tests/methods , Transcription, Genetic/drug effects , Valproic Acid/toxicity , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cluster Analysis , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Gene Expression Regulation/drug effects , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Principal Component Analysis , Risk Assessment
9.
Toxicol Appl Pharmacol ; 280(2): 378-88, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25150140

ABSTRACT

According to the advocated paradigm shift in toxicology, acquisition of knowledge on the mechanisms underlying the toxicity of chemicals, such as perturbations of biological pathways, is of primary interest. Pluripotent stem cells (PSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer a unique opportunity to derive physiologically relevant human cell types to measure molecular and cellular effects of such pathway modulations. Here we compared the neuronal differentiation propensity of hESCs and hiPSCs with the aim to develop novel hiPSC-based tools for measuring pathway perturbation in relation to molecular and cellular effects in vitro. Among other fundamental pathways, also, the cAMP responsive element binding protein (CREB) pathway was activated in our neuronal models and gave us the opportunity to study time-dependent effects elicited by chemical perturbations of the CREB pathway in relation to cellular effects. We show that the inhibition of the CREB pathway, using 2-naphthol-AS-E-phosphate (KG-501), induced an inhibition of neurite outgrowth and synaptogenesis, as well as a decrease of MAP2(+) neuronal cells. These data indicate that a CREB pathway inhibition can be related to molecular and cellular effects that may be relevant for neurotoxicity testing, and, thus, qualify the use of our hiPSC-derived neuronal model for studying chemical-induced neurotoxicity resulting from pathway perturbations.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Induced Pluripotent Stem Cells/cytology , Neurons/drug effects , Signal Transduction/drug effects , Cell Differentiation , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Embryonic Stem Cells/cytology , Gene Expression Profiling , Humans , Naphthols/pharmacology , Neurons/cytology , Organophosphates/pharmacology , Synapses/drug effects
10.
Stem Cells Transl Med ; 3(1): 54-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24307698

ABSTRACT

MicroRNAs (miRNAs) have recently been shown to act as regulatory signals for maintaining stemness and for determining the fate of adult and fetal stem cells, such as human mesenchymal stem cells (hMSCs). hMSCs constitute a population of multipotent stem cells that can be expanded easily in culture and are able to differentiate into many lineages. We have isolated two subpopulations of fetal mesenchymal stem cells (MSCs) from amniotic fluid (AF) known as spindle-shaped (SS) and round-shaped (RS) cells and characterized them on the basis of their phenotypes, pluripotency, proliferation rates, and differentiation potentials. In this study, we analyzed the miRNA profile of MSCs derived from AF, bone marrow (BM), and umbilical cord blood (UCB). We initially identified 67 different miRNAs that were expressed in all three types of MSCs but at different levels, depending on the source. A more detailed analysis revealed that miR-21 was expressed at higher levels in RS-AF-MSCs and BM-MSCs compared with SS-AF-MSCs. We further demonstrated for the first time a direct interaction between miR-21 and the pluripotency marker Sox2. The induction of miR-21 strongly inhibited Sox2 expression in SS-AF-MSCs, resulting in reduced clonogenic and proliferative potential and cell cycle arrest. Strikingly, the opposite effect was observed upon miR-21 inhibition in RS-AF-MSCs and BM-MSCs, which led to an enhanced proliferation rate. Finally, miR-21 induction accelerated osteogenesis and impaired adipogenesis and chondrogenesis in SS-AF-MSCs. Therefore, these findings suggest that miR-21 might specifically function by regulating Sox2 expression in human MSCs and might also act as a key molecule determining MSC proliferation and differentiation.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , MicroRNAs/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , 3' Untranslated Regions/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Homeodomain Proteins/genetics , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Transcriptome
11.
Expert Opin Drug Metab Toxicol ; 10(1): 39-50, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24102433

ABSTRACT

INTRODUCTION: Human stem cell lines and their derivatives, as alternatives to the use of animal cells or cancer cell lines, have been widely discussed as cellular models in predictive toxicology. However, the role of stem cells in the development of long-term toxicities and carcinogenesis has not received great attention so far, despite growing evidence indicating the relationship of stem cell damage to adverse effects later in life. However, testing this in vitro is a scientific/technical challenge in particular due to the complex interplay of factors existing under physiological conditions. Current major research programs in stem cell toxicity are not aiming to demonstrate that stem cells can be targeted by toxicants. Therefore, this knowledge gap needs to be addressed in additional research activities developing technical solutions and defining appropriate experimental designs. AREAS COVERED: The current review describes selected examples of the role of stem cells in the development of long-term toxicities in the brain, heart or liver and in the development of cancer. EXPERT OPINION: The presented examples illustrate the need to analyze the contribution of stem cells to chronic toxicity in order to make a final conclusion whether stem cell toxicities are an underestimated risk in mechanism-based safety assessments. This requires the development of predictive in vitro models allowing the assessment of adverse effects to stem cells on chronic toxicity and carcinogenicity.


Subject(s)
Stem Cells/drug effects , Stem Cells/pathology , Toxicity Tests, Chronic/methods , Animals , Brain/drug effects , Brain/metabolism , Carcinogens/toxicity , Cell Differentiation/drug effects , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Humans , Neoplasms/chemically induced , Neoplasms/pathology
12.
Arch Toxicol ; 87(1): 123-43, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23179753

ABSTRACT

Developmental neurotoxicity (DNT) and many forms of reproductive toxicity (RT) often manifest themselves in functional deficits that are not necessarily based on cell death, but rather on minor changes relating to cell differentiation or communication. The fields of DNT/RT would greatly benefit from in vitro tests that allow the identification of toxicant-induced changes of the cellular proteostasis, or of its underlying transcriptome network. Therefore, the 'human embryonic stem cell (hESC)-derived novel alternative test systems (ESNATS)' European commission research project established RT tests based on defined differentiation protocols of hESC and their progeny. Valproic acid (VPA) and methylmercury (MeHg) were used as positive control compounds to address the following fundamental questions: (1) Does transcriptome analysis allow discrimination of the two compounds? (2) How does analysis of enriched transcription factor binding sites (TFBS) and of individual probe sets (PS) distinguish between test systems? (3) Can batch effects be controlled? (4) How many DNA microarrays are needed? (5) Is the highest non-cytotoxic concentration optimal and relevant for the study of transcriptome changes? VPA triggered vast transcriptional changes, whereas MeHg altered fewer transcripts. To attenuate batch effects, analysis has been focused on the 500 PS with highest variability. The test systems differed significantly in their responses (<20 % overlap). Moreover, within one test system, little overlap between the PS changed by the two compounds has been observed. However, using TFBS enrichment, a relatively large 'common response' to VPA and MeHg could be distinguished from 'compound-specific' responses. In conclusion, the ESNATS assay battery allows classification of human DNT/RT toxicants on the basis of their transcriptome profiles.


Subject(s)
Embryonic Stem Cells/drug effects , Gene Expression Profiling , Mutagenicity Tests/methods , Neurotoxicity Syndromes/genetics , Binding Sites , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation/drug effects , Humans , Methylmercury Compounds/toxicity , Oligonucleotide Array Sequence Analysis , Valproic Acid/toxicity
13.
Stem Cells Dev ; 21(7): 1097-111, 2012 May 01.
Article in English | MEDLINE | ID: mdl-21988169

ABSTRACT

Recent studies support cell-based therapies for cancer treatment. An advantageous cell type for such therapeutic schemes are the mesenchymal stem cells (MSCs) that can be easily propagated in culture, genetically modified to express therapeutic proteins, and exhibit an innate tropism to solid tumors in vivo. Recently, we successfully isolated and expanded MSCs from second-trimester amniotic fluid (AF-MSCs). The main characteristic of AF-MSCs is their efficient and rapid expansion in vitro. Herein, we investigated the AF-MSCs tropism and capability to transport interferon beta (IFNß) to the region of neoplasia in a bladder tumor model. To this end, we used the T24M bladder cancer cell line, previously generated from our studies, and developed a disease progression model in immunosuppressed mice, that can recapitulate the molecular events of bladder carcinogenesis. Our results documented that AF-MSCs exhibited high motility, when migrated either to T24M cells or to T24M-conditioned medium, and we further identified and studied the secreted factors which may trigger these enhanced migratory properties. Further, lentivirus-transduced AF-MSCs, expressing green fluorescent protein (GFP) or IFNß, were intravenously administered to T24M tumor-bearing animals at multiple doses to examine their therapeutic effect. GFP- and IFNß-AF-MSCs successfully migrated and colonized at the tumor site. Notably, significant inhibition of tumor growth as well as prolonged survival of mice were observed in the presence of IFNß-AF-MSCs. Collectively, these results document the great potential of AF-MSCs as anti-cancer vehicles, implemented by the targeting of the tumor site and further facilitated by their high proliferation rate and expansion efficiency in culture.


Subject(s)
Amniotic Fluid/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Urinary Bladder Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Drug Delivery Systems , Green Fluorescent Proteins/biosynthesis , Humans , Interferon-beta/biosynthesis , Interferon-beta/blood , Interferon-beta/pharmacology , Interleukin-8/physiology , Kaplan-Meier Estimate , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Proteome/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/blood , Recombinant Proteins/pharmacology , Tumor Burden , Urinary Bladder Neoplasms/pathology , Vascular Endothelial Growth Factor A/physiology
14.
Gut ; 61(6): 894-906, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21997562

ABSTRACT

BACKGROUND: There is increasing interest in the therapeutic potential of human mesenchymal stem cells (hMSCs), especially in diseases such as acute hepatic failure (AHF) that are predominantly caused by a variety of drugs and viruses. In previous studies, a distinct population termed human spindle-shaped MSCs were isolated and expanded from second trimester amniotic fluid (AF-MSCs) and characterised based on their phenotype, pluripotency and differentiation potential. METHODS: AF-MSCs, hepatic progenitor-like (HPL) cells and hepatocyte-like (HL) cells derived from AF-MSCs were transplanted into CCl4-injured NOD/SCID mice with the AHF phenotype in order to evaluate their therapeutic potential. Conditioned medium (CM) derived from AF-MSCs or HPL cells was then delivered intrahepatically in order to determine whether the engraftment of the cells or their secreted molecules are the most important agents for liver repair. RESULTS: Both HPL cells and AF-MSCs were incorporated into CCl(4)-injured livers; HPL cell transplantation had a greater therapeutic effect. In contrast, HL cells failed to engraft and contribute to recovery. In addition, HPL-CM was found to be more efficient than CM derived from AF-MSCs in treatment of the liver. Proteome profile analysis of HPL-CM indicated the presence of anti-inflammatory factors such as interleukins IL-10, IL-1ra, IL-13 and IL-27 which may induce liver recovery. Blocking studies of IL-10 secretion from HPL cells confirmed the therapeutic significance of this cytokine in the AHF mouse model. CONCLUSIONS: Human spindle-shaped AF-MSCs or HPL cells might be valuable tools to induce liver repair and support liver function by cell transplantation. More importantly, the factors they release may also play an important role in cell treatment in diseases of the liver.


Subject(s)
Liver Failure, Acute/therapy , Mesenchymal Stem Cell Transplantation/methods , Amniotic Fluid/cytology , Animals , Green Fluorescent Proteins , Hepatocytes/cytology , Humans , In Situ Hybridization, Fluorescence , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-2/blood , Mesenchymal Stem Cells/physiology , Mice , Protein Array Analysis , Tumor Necrosis Factor-alpha/blood
15.
J Cell Mol Med ; 15(9): 1896-913, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21166769

ABSTRACT

Human mesenchymal progenitor cells (MPCs) are considered to be of great promise for use in tissue repair and regenerative medicine. MPCs represent multipotent adherent cells, able to give rise to multiple mesenchymal lineages such as osteoblasts, adipocytes or chondrocytes. Recently, we identified and characterized human second trimester amniotic fluid (AF) as a novel source of MPCs. Herein, we found that early colonies of AF-MPCs consisted of two morphologically distinct adherent cell types, termed as spindle-shaped (SS) and round-shaped (RS). A detailed analysis of these two populations showed that SS-AF-MPCs expressed CD90 antigen in a higher level and exhibited a greater proliferation and differentiation potential. To characterize better the molecular identity of these two populations, we have generated a comparative proteomic map of SS-AF-MPCs and RS-AF-MPCs, identifying 25 differentially expressed proteins and 10 proteins uniquely expressed in RS-AF-MPCs. Furthermore, SS-AF-MPCs exhibited significantly higher migration ability on extracellular matrices, such as fibronectin and laminin in vitro, compared to RS-AF-MPCs and thus we further evaluated SS-AF-MPCs for potential use as therapeutic tools in vivo. Therefore, we tested whether GFP-lentiviral transduced SS-AF-MPCs retained their stem cell identity, proliferation and differentiation potential. GFP-SS-AF-MPCs were then successfully delivered into immunosuppressed mice, distributed in different tissues and survived longterm in vivo. In summary, these results demonstrated that AF-MPCs consisted of at least two different MPC populations. In addition, SS-AF-MPCs, isolated based on their colony morphology and CD90 expression, represented the only MPC population that can be expanded easily in culture and used as an efficient tool for future in vivo therapeutic applications.


Subject(s)
Amniotic Fluid/cytology , Mesenchymal Stem Cells/cytology , Animals , Antibodies, Neutralizing/pharmacology , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Electrophoresis, Gel, Two-Dimensional , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibronectins/pharmacology , Humans , Hyaluronan Receptors/immunology , Hyaluronic Acid/pharmacology , Integrin alpha5/metabolism , Lentivirus/drug effects , Lentivirus/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Phenotype , Reproducibility of Results , Thy-1 Antigens/metabolism , Transcription Factors/metabolism , Transduction, Genetic
16.
Stem Cells Dev ; 16(6): 931-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18047393

ABSTRACT

Human mesenchymal stem cells (hMSCs) constitute a population of multipotent adherent cells able to give rise to multiple mesenchymal lineages such as osteoblasts, adipocytes, or chondrocytes. So far, the most common source of MSCs has been the bone marrow (BM); however BM-MSC harvesting and processing exhibits major drawbacks and limitations. Thus, identification and characterization of alternative sources of MSCs are of great importance. In the present study, we isolated and expanded fetal MSCs from second-trimester amniotic fluid (AF). We documented that these cells are of embryonic origin, can differentiate under appropriate conditions into cell types derived from all three germ layers, and express the pluripotency marker Oct-4, the human Nanog protein, and the stage-specific embryonic antigen-4 (SSEA-4). Furthermore, we systematically tested the immunophenotype of cultured MSCs by flow cytometry analysis using a wide variety of markers. Direct comparison of this phenotype to the one derived from cultured BM-MSCs demonstrated that cultured MSCs from both sources exhibit similar expression patterns. Using the two-dimensional gel electrophoresis and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) approach, we have generated for the first time the protein map of cultured AF-MSCs by identifying 261 proteins, and we compared it directly to that of cultured BM-MSCs. The functional pattern of the identified proteins from both sources was similar. However, cultured AF-MSCs displayed a number of unique proteins related to proliferation and primitive phenotype, which may confer to the distinct features of the two types. Considering the easy access to this new cell source and the yield of expanded MSCs for stem cell research, AF may provide an excellent source of MSCs both for basic research and for potential therapeutic applications.


Subject(s)
Amniotic Fluid/cytology , Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Proteomics , Amniocentesis , Amniotic Fluid/physiology , Antigens, CD/analysis , Antigens, CD/genetics , Bone Marrow Cells/physiology , Cell Culture Techniques , Cell Differentiation , Cell Division , Female , Humans , Kinetics , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...