Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Haematologica ; 100(1): 32-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326431

ABSTRACT

Epigenetic memory in induced pluripotent stem cells, which is related to the somatic cell type of origin of the stem cells, might lead to variations in the differentiation capacities of the pluripotent stem cells. In this context, induced pluripotent stem cells from human CD34(+) hematopoietic stem cells might be more suitable for hematopoietic differentiation than the commonly used fibroblast-derived induced pluripotent stem cells. To investigate the influence of an epigenetic memory on the ex vivo expansion of induced pluripotent stem cells into erythroid cells, we compared induced pluripotent stem cells from human neural stem cells and human cord blood-derived CD34(+) hematopoietic stem cells and evaluated their potential for differentiation into hematopoietic progenitor and mature red blood cells. Although genome-wide DNA methylation profiling at all promoter regions demonstrates that the epigenetic memory of induced pluripotent stem cells is influenced by the somatic cell type of origin of the stem cells, we found a similar hematopoietic induction potential and erythroid differentiation pattern of induced pluripotent stem cells of different somatic cell origin. All human induced pluripotent stem cell lines showed terminal maturation into normoblasts and enucleated reticulocytes, producing predominantly fetal hemoglobin. Differences were only observed in the growth rate of erythroid cells, which was slightly higher in the induced pluripotent stem cells derived from CD34(+) hematopoietic stem cells. More detailed methylation analysis of the hematopoietic and erythroid promoters identified similar CpG methylation levels in the induced pluripotent stem cell lines derived from CD34(+) cells and those derived from neural stem cells, which confirms their comparable erythroid differentiation potential.


Subject(s)
Cell Differentiation , Erythroid Cells/cytology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Biomarkers/metabolism , DNA Methylation , Epigenomics , Erythroid Cells/metabolism , Fetal Blood/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
2.
Cancer Biomark ; 14(5): 295-302, 2014.
Article in English | MEDLINE | ID: mdl-25171471

ABSTRACT

Urinary DNA is increasingly gaining importance in diagnosis of urological malignancies. Especially cell-free DNA originating from apoptotic and necrotic cells of the early tumor could become a key target for early stage tumor diagnosis. Aberrant DNA methylation forms tumor cell characteristic epigenetic profiles which are covalently established before any tumor related aberration at transcriptional or protein level has occurred. In addition, these epigenetic signatures are alterably adapted to and accompanying the individual stages of multistep, progressive tumorigenesis. Hence, they seem very promising for diagnosis as well as for monitoring the patient's follow-up care and even for decisions regarding personalized therapeutic options. The essential prerequisite at this approach will be a reliable methodological handling of the biological material of interest. In this study we present detailed analyses of LINE-1 DNA methylation profiles and demonstrate the sensitive detection of LINE-1 DNA methylation differences as well as between cancer patients and healthy individuals, between urinary cellular and cell-free DNA. In addition, we show methylome differences between both DNA fractions from a healthy individual and bladder cancer patients. In conclusion, we demonstrate here the unrestricted amenability of urinary cell-free DNA for both, a detailed characterization of a distinct DNA methylation alteration and its sensitive detection and a comprehensive global, array-based screening for DNA methylation differences.


Subject(s)
Cell-Free System/physiology , DNA Methylation/genetics , DNA/genetics , Epigenesis, Genetic/genetics , Urinary Bladder/physiology , Case-Control Studies , Humans , Urinary Bladder Neoplasms/genetics
3.
PLoS One ; 8(3): e59011, 2013.
Article in English | MEDLINE | ID: mdl-23527072

ABSTRACT

MicroRNAs are key regulators of neural cell proliferation, differentiation and fate choice. Due to the limited access to human primary neural tissue, the role of microRNAs in human neuronal differentiation remains largely unknown. Here, we use a population of long-term self-renewing neuroepithelial-like stem cells (lt-NES cells) derived from human embryonic stem cells to study the expression and function of microRNAs at early stages of human neural stem cell differentiation and neuronal lineage decision. Based on microRNA expression profiling followed by gain- and loss-of-function analyses in lt-NES cells and their neuronal progeny, we demonstrate that miR-153, miR-324-5p/3p and miR-181a/a contribute to the shift of lt-NES cells from self-renewal to neuronal differentiation. We further show that miR-125b and miR-181a specifically promote the generation of neurons of dopaminergic fate, whereas miR-181a inhibits the development of this neurotransmitter subtype. Our data demonstrate that time-controlled modulation of specific microRNA activities not only regulates human neural stem cell self-renewal and differentiation but also contributes to the development of defined neuronal subtypes.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , Neurons/cytology , Neurons/metabolism , Cell Lineage/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurites/metabolism , Reproducibility of Results
4.
BMC Genomics ; 14: 111, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23418963

ABSTRACT

BACKGROUND: MicroRNAs are a population of short non-coding RNAs with widespread negative regulatory impact on mRNA translation. Unrestricted somatic stem cells (USSC) are a rare population in human cord blood that can be induced into cells representative of all three germinal layers. Here we analyzed the functional impact of miRNAs on the osteogenic differentiation in USSC. RESULTS: Gene expression profiling identified 20 microRNAs that were consistently upregulated during osteogenic differentiation of two different USSC cell lines (SA5/73 and SA8/25). Bioinformatic target gene prediction indicated that among these microRNAs, miR-10a, -22, -26a, -26b, and -29b recognize transcripts that encode a set of proteins inhibiting osteogenesis. We subsequently verified osteo-inhibitory CDK6, CTNNBIP1, HDAC4, and TOB1 and osteo-promoting SMAD1 as targets of these microRNAs. In Western blot analyses demonstrated that endogenous levels of CDK6 and HDAC4 were downregulated during osteogenic differentiation of USSC and reduced following ectopic expression of miR-26a/b and miR-29b. In contrast, endogenous expression of SMAD1, targeted by miR-26a/b, was unaltered during osteogenic differentiation of USSC or following ectopic expression of miR-26a/b. Functional overexpression analyses using microRNA mimics revealed that miR-26a/b, as well as miR-29b strongly accelerated osteogenic differentiation of USSC as assessed by Alizarin-Red staining and calcium-release assays. CONCLUSIONS: miR-26a/b and miR-29b are upregulated during osteogenic differentiation of USSC and share target genes inhibiting osteogenesis. Furthermore, these microRNAs accelerate osteogenic differentiation, likely mediated by osteo-inhibitory proteins such as CDK6 and HDAC4.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation/genetics , Fetal Blood/cytology , MicroRNAs/genetics , Osteogenesis/genetics , Adaptor Proteins, Signal Transducing , Adult Stem Cells/metabolism , Computational Biology , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Fetal Blood/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcriptome/genetics , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
6.
Nat Methods ; 9(6): 575-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484851

ABSTRACT

Forced expression of proneural transcription factors has been shown to direct neuronal conversion of fibroblasts. Because neurons are postmitotic, conversion efficiencies are an important parameter for this process. We present a minimalist approach combining two-factor neuronal programming with small molecule-based inhibition of glycogen synthase kinase-3ß and SMAD signaling, which converts postnatal human fibroblasts into functional neuron-like cells with yields up to >200% and neuronal purities up to >80%.


Subject(s)
Cell Transdifferentiation , Fibroblasts/physiology , Neurons/physiology , Child, Preschool , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Infant , Infant, Newborn , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Transcription Factors/pharmacology
7.
Brain ; 135(Pt 2): 431-46, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21903726

ABSTRACT

Stem cell therapy is a potential treatment for spinal cord injury and different stem cell types have been grafted into animal models and humans suffering from spinal trauma. Due to inconsistent results, it is still an important and clinically relevant question which stem cell type will prove to be therapeutically effective. Thus far, stem cells of human sources grafted into spinal cord mostly included barely defined heterogeneous mesenchymal stem cell populations derived from bone marrow or umbilical cord blood. Here, we have transplanted a well-defined unrestricted somatic stem cell isolated from human umbilical cord blood into an acute traumatic spinal cord injury of adult immune suppressed rat. Grafting of unrestricted somatic stem cells into the vicinity of a dorsal hemisection injury at thoracic level eight resulted in hepatocyte growth factor-directed migration and accumulation within the lesion area, reduction in lesion size and augmented tissue sparing, enhanced axon regrowth and significant functional locomotor improvement as revealed by three behavioural tasks (open field Basso-Beattie-Bresnahan locomotor score, horizontal ladder walking test and CatWalk gait analysis). To accomplish the beneficial effects, neither neural differentiation nor long-lasting persistence of the grafted human stem cells appears to be required. The secretion of neurite outgrowth-promoting factors in vitro further suggests a paracrine function of unrestricted somatic stem cells in spinal cord injury. Given the highly supportive functional characteristics in spinal cord injury, production in virtually unlimited quantities at GMP grade and lack of ethical concerns, unrestricted somatic stem cells appear to be a highly suitable human stem cell source for clinical application in central nervous system injuries.


Subject(s)
Cord Blood Stem Cell Transplantation , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Axons/physiology , Cells, Cultured , Female , Humans , Motor Activity/physiology , Rats , Rats, Wistar , Spinal Cord Injuries/physiopathology
8.
PLoS One ; 6(1): e16138, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21283765

ABSTRACT

BACKGROUND: MicroRNAs are short (∼22 nt) non-coding regulatory RNAs that control gene expression at the post-transcriptional level. Here the functional impact of microRNAs on cell cycle arrest during neuronal lineage differentiation of unrestricted somatic stem cells from human cord blood (USSC) was analyzed. METHODOLOGY/PRINCIPAL FINDINGS: Expression profiling revealed downregulation of microRNAs miR-17, -20a, and -106b in USSC differentiated into neuronal lineage but not in USSC differentiated into osteogenic lineage. Transfection experiments followed by Ki67 immunostainings demonstrated that each of these microRNAs was able to promote proliferation of native USSC and to prevent in part cell cycle arrest during neuronal lineage differentiation of USSC. Bioinformatic target gene predictions followed by experimental target gene validations revealed that miR-17, -20a, and -106b act in a common manner by downregulating an overlapping set of target genes mostly involved in regulation and execution of G(1)/S transition. Pro-proliferative target genes cyclinD1 (CCND1) and E2F1 as well as anti-proliferative targets CDKN1A (p21), PTEN, RB1, RBL1 (p107), RBL2 (p130) were shown as common targets for miR-17, -20a, and -106b. Furthermore, these microRNAs also downregulate WEE1 which is involved in G(2)/M transition. Most strikingly, miR-17, -20a, and -106b were found to promote cell proliferation by increasing the intracellular activity of E2F transcription factors, despite the fact that miR-17, -20a, and -106b directly target the transcripts that encode for this protein family. CONCLUSIONS/SIGNIFICANCE: Mir-17, -20a, and -106b downregulate a common set of pro- and anti-proliferative target genes to impact cell cycle progression of USSC and increase intracellular activity of E2F transcription factors to govern G(1)/S transition.


Subject(s)
Cell Cycle , Cell Lineage , E2F Transcription Factors/metabolism , Fetal Blood/cytology , MicroRNAs/physiology , Neurons/cytology , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Down-Regulation , G1 Phase/genetics , Humans , Stem Cells/cytology
9.
Stem Cell Res ; 6(1): 60-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20933485

ABSTRACT

Unrestricted somatic stem cells (USSC) from human cord blood display a broad differentiation potential for ectodermal, mesodermal, and endodermal cell types. The molecular basis for these stem cell properties is unclear and unlike embryonic stem cells (ESC) none of the major stem cell factors OCT4, SOX2, and NANOG exhibits significant expression in USSC. Here, we report that these key stem cell genes hold an epigenetic state in between that of an ESC and a terminally differentiated cell type. DNA methylation analysis exhibits partial demethylation of the regulatory region of OCT4 and a demethylated state of the NANOG and SOX2 promoter/enhancer regions. Further genome-wide DNA methylation profiling identified a partially demethylated state of the telomerase gene hTERT. Moreover, none of the pluripotency factors exhibited a repressive histone signature. Notably, SOX2 exhibits a bivalent histone signature consisting of the opposing histone marks dimeH3K4 and trimeH3K27, which is typically found on genes that are "poised" for transcription. Consequently, ectopic expression of OCT4 in USSC led to rapid induction of expression of its known target gene SOX2. Our data suggest that incomplete epigenetic repression and a "poised" epigenetic status of pluripotency genes preserves the USSC potential to be able to react adequately to distinct differentiation and reprogramming cues.


Subject(s)
Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Fetal Blood/cytology , Gene Expression Regulation, Developmental , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , DNA Methylation , Embryonic Stem Cells/cytology , Female , Fetal Blood/metabolism , Humans , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism
10.
Stem Cells Dev ; 20(8): 1383-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21067317

ABSTRACT

Unrestricted somatic stem cells (USSCs) represent an intrinsically multipotent CD45-negative fetal population from human cord blood. They show differentiation into neuronal cells of a dopaminergic phenotype, which express neuronal markers such as synaptophysin, neuronal-specific nuclear protein, and neurofilament and release the neurotransmitter dopamine accompanied by expression of dopaminergic key factors tyrosine hydroxylase and Nurr1 (NR4A2). MicroRNA expression analysis highlighted their importance in neural development but their specific functions remain poorly understood. Here, downregulation of a set of 18 microRNAs during neuronal lineage differentiation of unrestricted somatic stem cells, including members of the miR-17-92 family and additional microRNAs such as miR-130a, -138, -218, and -335 as well as their target genes, is described. In silico target gene predictions for this microRNA group uncovered a large set of proteins involved in neuronal differentiation and having a strong impact on differentiation-related pathways such as axon guidance and TGFß, WNT, and MAPK signaling. Experimental target validations confirmed approximately 35% of predictions tested and revealed a group of proteins with specific impact on neuronal differentiation and function including neurobeachin, neurogenic differentiation 1, cysteine-rich motor neuron protein 1, neuropentraxin 1, and others. These proteins are combined targets for several subgroups from the set of 18 downregulated microRNAs. This finding was further supported by the observed upregulation of a significant amount of predicted and validated target genes based on Illumina Beadstudio microarray data. Confirming the functional relationship of a limited panel of microRNAs and predicted target proteins reveals a clear network-like impact of the group of 18 downregulated microRNAs on proteins involved in neuronal development and function.


Subject(s)
Cell Lineage/genetics , MicroRNAs/metabolism , Neurogenesis/genetics , Stem Cells/cytology , Cell Line , Dopamine/biosynthesis , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/metabolism , Neurofilament Proteins/biosynthesis , Neurons/metabolism , Nuclear Proteins/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Stem Cells/metabolism , Synaptophysin/biosynthesis , Transforming Growth Factor beta/metabolism , Tyrosine 3-Monooxygenase/biosynthesis , Wnt Proteins/metabolism
11.
Exp Hematol ; 38(11): 1099-104, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20869422

ABSTRACT

OBJECTIVE: Generation and expression of unrestricted somatic stem cells (USSC) from human cord blood as well as their in vitro functional characterization at the clonal level. MATERIALS AND METHODS: USSC generation was initiated from fresh cord blood followed by lentiviral transfection and clone generation via limiting dilution. Individual clones were analyzed for lentiviral genomic integration patterns by ligation-mediated polymerase chain reaction. In vitro differentiation of clonal USSC was performed into mesodermal, endodermal, and ectodermal lineages according to our published protocols. Respective osteogenic, hepatic, and neuronal lineage-specification was documented by immunohistochemistry and tissue-specific protein expression was analyzed by Western blotting. MicroRNA expression analysis was achieved using the TaqMan microRNA Megaplex array. RESULTS: Lentivirally labeled USSC cultures were successfully subjected to limiting dilution cloning. One clone containing a single lentiviral integration site was identified (clone 4) and used for further differentiation experiments. Ligation-mediated polymerase chain reaction results from mesodermally, endodermally, and ectodermally differentiated USSC clone 4 consistently showed only the primary single lentiviral integration site. Lineage-specific differentiation experiments were confirmed by morphology and cell-fate-specific monoclonal antibodies in immunocytochemistry. MicroRNA expression profiles did not reveal dramatic differences between clonal and nonclonal USSC. CONCLUSIONS: The proof of the clonal existence of USSC is important for the assessment of biological properties unique for these unrestricted human stem cell candidates. As clones they can be subjected to advanced methods that enable defining of the multilayer nature of regulatory mechanisms through single-cell analysis.


Subject(s)
Cell Differentiation , Fetal Blood/cytology , Germ Layers/cytology , Stem Cells/cytology , Blotting, Western , Cell Lineage , Clone Cells/cytology , Clone Cells/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Endoderm/cytology , Endoderm/metabolism , Gene Expression Profiling , Germ Layers/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Lac Operon/genetics , Lentivirus/genetics , Mesoderm/cytology , Mesoderm/metabolism , MicroRNAs/genetics , Neurons/cytology , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Osteoblasts/cytology , Osteoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Transduction, Genetic , beta-Galactosidase/metabolism
12.
BMC Res Notes ; 3: 219, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20682048

ABSTRACT

BACKGROUND: The miR-200c/141 cluster has recently been implicated in the epithelial to mesenchymal transition (EMT) process. The expression of these two miRNAs is inversely correlated with tumorigenicity and invasiveness in several human cancers. The role of these miRNAs in cancer progression is based in part on their capacity to target the EMT activators ZEB1 and ZEB2, two transcription factors, which in turn repress expression of E-cadherin. Little is known about the regulation of the mir200c/141 cluster, whose targeting has been proposed as a promising new therapy for the most aggressive tumors. FINDINGS: We show that the miR-200c/141 cluster is repressed by DNA methylation of a CpG island located in the promoter region of these miRNAs. Whereas in vitro methylation of the miR-200c/141 promoter led to shutdown of promoter activity, treatment with a demethylating agent caused transcriptional reactivation in breast cancer cells formerly lacking expression of miR-200c and miR-141. More importantly, we observed that DNA methylation of the identified miR-200c/141 promoter was tightly correlated with phenotype and the invasive capacity in a panel of 8 human breast cancer cell lines. In line with this, in vitro induction of EMT by ectopic expression of the EMT transcription factor Twist in human immortalized mammary epithelial cells (HMLE) was accompanied by increased DNA methylation and concomitant repression of the miR-200c/141 locus. CONCLUSIONS: The present study demonstrates that expression of the miR-200c/141 cluster is regulated by DNA methylation, suggesting epigenetic regulation of this miRNA locus in aggressive breast cancer cell lines as well as untransformed mammary epithelial cells. This epigenetic silencing mechanism might represent a novel component of the regulatory circuit for the maintenance of EMT programs in cancer and normal cells.

13.
Exp Hematol ; 38(9): 809-18, 818.e1-2, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20541586

ABSTRACT

OBJECTIVE: Generation of induced pluripotent stem (iPS) cells from human cord blood (CB)-derived unrestricted somatic stem cells and evaluation of their molecular signature and differentiation potential in comparison to human embryonic stem cells. MATERIALS AND METHODS: Unrestricted somatic stem cells isolated from human CB were reprogrammed to iPS cells using retroviral expression of the transcription factors OCT4, SOX2, KLF4, and C-MYC. The reprogrammed cells were analyzed morphologically, by quantitative reverse transcription polymerase chain reaction, genome-wide microRNA and methylation profiling, and gene expression microarrays, as well as in their pluripotency potential by in vivo teratoma formation in severe combined immunodeficient mice and in vitro differentiation. RESULTS: CB iPS cells are very similar to human embryonic stem cells morphologically, at their molecular signature, and in their differentiation potential. CONCLUSIONS: Human CB-derived unrestricted somatic stem cells offer an attractive source of cells for generation of iPS cells. Our findings open novel perspectives to generate human leukocyte antigen-matched pluripotent stem cell banks based on existing CB banks. Besides the obvious relevance of a second-generation CB iPS cell bank for pharmacological and toxicological testing, its application for autologous or allogenic regenerative cell transplantation appears feasible.


Subject(s)
Cell Dedifferentiation , Fetal Blood/cytology , Fetal Blood/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , DNA Methylation/genetics , Genome-Wide Association Study , Humans , Kruppel-Like Factor 4 , Mice , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Stem Cell Transplantation , Teratoma/metabolism , Teratoma/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transplantation, Autologous , Transplantation, Heterologous , Transplantation, Homologous
14.
Stem Cells Dev ; 19(10): 1471-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20331358

ABSTRACT

In addition to hematopoietic stem cells, cord blood (CB) also contains different nonhematopoietic CD45-, CD34- adherent cell populations: cord blood mesenchymal stromal cells (CB MSC) that behave almost like MSC from bone marrow (BM MSC) and unrestricted somatic stem cells (USSC) that differentiate into cells of all 3 germ layers. Distinguishing between these populations is difficult due to overlapping features such as the immunophenotype or the osteogenic and chondrogenic differentiation pathway. Functional differences in the differentiation potential suggest different developmental stages or different cell populations. Here we demonstrate that the expression of genes and the differentiation toward the adipogenic lineage can discriminate between these 2 populations. USSC, including clonal-derived cells lacking adipogenic differentiation, strongly expressed δ-like 1/preadipocyte factor 1 (DLK-1/PREF1) correlating with high proliferative potential, while CB MSC were characterized by a strong differentiation toward adipocytes correlating with a weak or negative DLK-1/PREF1 expression. Constitutive overexpression of DLK-1/PREF1 in CB MSC resulted in a reduced adipogenic differentiation, whereas silencing of DLK-1 in USSC resulted in adipogenic differentiation.


Subject(s)
Biomarkers/metabolism , Cell Differentiation , Fetal Blood/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Stem Cells/metabolism , Stromal Cells/metabolism , Calcium-Binding Proteins , Gene Expression Regulation , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Stem Cells/cytology , Stromal Cells/cytology
15.
J Cell Mol Med ; 13(8B): 2465-2475, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19175687

ABSTRACT

Unrestricted somatic stem cells (USSC) have the potential to differentiate into tissues derived from all three germinal layers and therefore hold promise for use in regenerative therapies. Furthermore, they have haematopoietic stromal activity, a characteristic that may be exploited to enhance haematopoietic engraftment. Both applications may require USSC to be used in an allogeneic, HLA-mismatched setting. We have therefore studied their in vitro interaction with cellular immunity. USSC showed no allostimulatory activity and caused only minimal inhibition of allogeneic T-cell responses. However, following pre-stimulation with IFNgamma and TNFalpha, they inhibited T-cell proliferation in an indoleamine 2, 3-dioxygenase-dependent manner and suppressed graft-versus-host type reactions. In addition, USSC inhibited DC maturation and function. This inhibition was overridden by stronger DC maturation signals provided by IL-1beta, IL-6, PGE(2) and TNFalpha compared to TNFalpha alone. Pre-stimulation of USSC with IFNgamma and TNFalpha had a similar effect: Inhibition of DC maturation was no longer observed. Thus, USSC are conditionally immunosuppressive, and IFNgamma and TNFalpha constitute a switch, which regulates their immunological properties. They either suppress T-cell responses in the presence of both cytokines or in their absence block DC differentiation and function. These activities may contribute to fine-tuning the immune system especially at sites of tissue damage in order to ensure appropriate differentiation of USSC and subsequent tissue repair. Therapeutically, they may help to protect USSC and possibly their progeny from immune rejection.


Subject(s)
Cytokines/physiology , Fetal Blood , Immunity, Cellular , Stem Cells/cytology , Flow Cytometry , Graft vs Host Reaction , Humans , Immunophenotyping , Lymphocyte Culture Test, Mixed
16.
J Biol Chem ; 283(47): 32244-53, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18628208

ABSTRACT

An under-agarose chemotaxis assay was used to investigate whether unrestricted somatic stem cells (USSC) that were recently characterized in human cord blood are attracted by neuronal injury in vitro. USSC migrated toward extracts of post-ischemic brain tissue of mice in which stroke had been induced. Moreover, apoptotic neurons secrete factors that strongly attracted USSC, whereas necrotic and healthy neurons did not. Investigating the expression of growth factors and chemokines in lesioned brain tissue and neurons and of their respective receptors in USSC revealed expression of hepatocyte growth factor (HGF) in post-ischemic brain and in apoptotic but not in necrotic neurons and of the HGF receptor c-MET in USSC. Neuronal lesion-triggered migration was observed in vitro and in vivo only when c-MET was expressed at a high level in USSC. Neutralization of the bioactivity of HGF with an antibody inhibited migration of USSC toward neuronal injury. This, together with the finding that human recombinant HGF attracts USSC, document that HGF signaling is necessary for the tropism of USSC for neuronal injury. Our data demonstrate that USSC have the capacity to migrate toward apoptotic neurons and injured brain. Together with their neural differentiation potential, this suggests a neuroregenerative potential of USSC. Moreover, we provide evidence for a hitherto unrecognized pivotal role of the HGF/c-MET axis in guiding stem cells toward brain injury, which may partly account for the capability of HGF to improve function in the diseased central nervous system.


Subject(s)
Fetal Blood/metabolism , Hepatocyte Growth Factor/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-met/metabolism , Stem Cells/cytology , Animals , Apoptosis , Brain/metabolism , Brain Ischemia/pathology , Cell Movement , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological
17.
Stem Cells Dev ; 17(2): 221-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18447638

ABSTRACT

Recently, it has been shown that human unrestricted somatic stem cells (USSCs) from umbilical cord blood represent pluripotent, neonatal, nonhematopoietic stem cells with the potential to differentiate into the neural lineage. However, molecular and functional characterization of the neural phenotype and evaluation of the degree of maturity of the resulting cells are still lacking. In this study, we addressed the question of neuronal differentiation and maturation induced by a defined composition of growth and differentiation factors (XXL medium). We demonstrated the expression of different neuronal markers and their enrichment in USSC cultures during XXL medium incubation. Furthermore, we showed enrichment of USSCs expressing tyrosine hydroxylase (TH), an enzyme specific for dopaminergic neurons and other catecholamine-producing neurons, accompanied by induction of Nurr1, a factor regulating dopaminergic neurogenesis. The functionality of USSCs has been analyzed by patch-clamp recordings and high-performance liquid chromatography (HPLC). Voltage-gated sodium-channels could be identified in laminin-predifferentiated USSCs. In addition, HPLC analysis revealed synthesis and release of the neurotransmitter dopamine by USSC-derived cells, thus correlating well with the detection of TH transcripts and protein. This study provides novel insight into the potential of unrestricted somatic stem cells from human umbilical cord blood to acquire a neuronal phenotype and function.


Subject(s)
Cell Differentiation , Dopamine/metabolism , Fetal Blood/cytology , Neurons/physiology , Stem Cells/physiology , Animals , Cell Cycle/drug effects , Cells, Cultured , Culture Media/pharmacology , Humans , Neurons/metabolism , Phenotype , Rats , Sodium Channels/metabolism , Sodium Channels/physiology , Stem Cells/metabolism
18.
Haematologica ; 93(3): 347-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18268278

ABSTRACT

BACKGROUND: Pegylated granulocyte colony-stimulating factor (G-CSF) has recently been introduced as a new compound for mobilization of CD34(+) hematopoietic stem and progenitor cells. In this study, we compared the molecular and functional characteristics of CD34(+) cells mobilized by pegylated G-CSF with those mobilized by unconjugated G-CSF. DESIGN AND METHODS: Gene expression of immunomagnetically enriched CD34(+) cells from leukapheresis products of patients who were given pegylated-G-CSF or unconjugated G-CSF was analyzed using Affymetrix HG Focus microarrays and quantitative reverse transcriptase polymerase chain reaction. Flow cytometry and fluorescence activated cell sorting was conducted to assess the CD34(+) subset composition and to obtain Lin(-), CD34(+), CD38(-) hematopoietic stem cells. Cell cycle assays and clonogenic assays were performed for functional corroboration. RESULTS: Pegylated G-CSF and unconjugated G-CSF mobilized CD34(+) and hematopoietic stem cells with different molecular phenotypes and functional properties. The CD34(+) cells mobilized by pegylated G-CSF had higher expression levels of genes indicative of early hematopoiesis, including HOXA9, MEIS1 and GATA3. We found lower expression of genes characteristic of erythroid and later stages of myeloid differentiation and a lower functional burst-forming unit erythroid/colony-forming unit-granulocyte-macrophage ratio. Consistently, greater numbers of hematopoietic stem cells and common myeloid progenitors and fewer megakaryocyte-erthrocyte progenitors were found in the pegylated-G-CSF-mobilized CD34(+) cells. Additionally, sorted pegylated-G-CSF-mobilized hematopoietic stem cells displayed higher expression of HOXA9 in comparison to G-CSF-mobilized hematopoietic stem cells. In line with the gene expression data, CD34(+) cells mobilized by pegylated G-CSF, as well as sorted hematopoietic stem cells, showed a significantly greater cell cycle activity. CONCLUSIONS: Stimulation with pegylated-G-CSF or G-CSF results in different expression of key regulatory genes and different functional properties of mobilized hematopoietic stem cells as well as their progeny, a finding that might be relevant for the application of these cells in blood stem cell transplantation.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Adult , Aged , Antigens, CD34/analysis , Cell Cycle , Cell Division/genetics , Colony-Forming Units Assay , Female , Filgrastim , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoiesis/genetics , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/cytology , Humans , Immunomagnetic Separation , Leukapheresis , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polyethylene Glycols , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction
19.
Cell Signal ; 19(12): 2428-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17651940

ABSTRACT

Self-renewing divisions of normal and cancerous stem cells are responsible for the initiation and maintenance of normal and certain cancerous tissues, respectively. Recent findings suggest that tumor surveillance mechanisms can reduce regenerative capacity and frequency of normal stem cells, thereby contributing to tissue aging. Signaling pathways promoting self-renewal of stem cells can also drive proliferation in cancer. The BMI-1 proto-oncogene is required for the maintenance of tissue-specific stem cells and is involved in carcinogenesis within the same tissues. BMI-1 promotes self-renewal of stem cells largely by interfering with two central cellular tumor suppressor pathways, p16(Ink4a)/retinoblastoma protein (Rb) and ARF/p53, whose disruption is a hallmark of cancer. Nucleolin, an Rb-associated protein, is abundant in proliferating cancerous cells and likely contributes to the maintenance of human CD34-positive stem/progenitor cells of hematopoiesis. Elucidation of the involvement of proto-oncogenes and tumor suppressors in the maintenance of stem cells might have therapeutic implications.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Stem Cells/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Neoplasms/pathology , Phosphoproteins/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Nucleolin
20.
Cell ; 129(7): 1401-14, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17604727

ABSTRACT

MicroRNAs (miRNAs) are small noncoding regulatory RNAs that reduce stability and/or translation of fully or partially sequence-complementary target mRNAs. In order to identify miRNAs and to assess their expression patterns, we sequenced over 250 small RNA libraries from 26 different organ systems and cell types of human and rodents that were enriched in neuronal as well as normal and malignant hematopoietic cells and tissues. We present expression profiles derived from clone count data and provide computational tools for their analysis. Unexpectedly, a relatively small set of miRNAs, many of which are ubiquitously expressed, account for most of the differences in miRNA profiles between cell lineages and tissues. This broad survey also provides detailed and accurate information about mature sequences, precursors, genome locations, maturation processes, inferred transcriptional units, and conservation patterns. We also propose a subclassification scheme for miRNAs for assisting future experimental and computational functional analyses.


Subject(s)
Base Sequence/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Library , MicroRNAs/genetics , Animals , Cell Lineage/genetics , Conserved Sequence/genetics , Hematologic Neoplasms/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mice , Molecular Sequence Data , Phylogeny , RNA, Messenger/genetics , Rats , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...