Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Bioengineering (Basel) ; 11(3)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38534553

ABSTRACT

BACKGROUND: The lack of resources limits the treatment of craniomaxillofacial fractures (CMF) in low-income countries (LIC). Therefore, Barton bandages and/or interdental wiring are considered in these regions. Fracture reduction is maintained by permanent occlusion for 6 weeks, which often leads to limited compliance and dissatisfying results. The aim of this cadaver-based study is to evaluate the feasibility of the use of an external face fixator (EFF) for the treatment of CMF, its biomechanical values and to define the optimal pin insertion points and angles. MATERIALS AND METHODS: An AO hand fixator was used. CMF of types Le Fort 1-3 with split fractures of the hard palate were treated with EFF on 13 anatomical specimens. Fractures were created using a chisel, and pins were placed in specific anatomical regions. The maximal pull-out force [N] of pins was analysed by a tensile force gauge, and Fmax of the mandibular pins was evaluated. Computer tomography scans were performed on the healthy, fractured and EFF-treated skulls. RESULTS: The pull-out forces for the single pins were mandibular pins (n = 15, median 488.0 N), supraorbital pins (n = 15, median 455.0 N), zygomatic pins (n = 14, median 269.1 N), medial hard palate pins (n = 12, median 208.4 N) and lateral hard palate pins (n = 8, median 49.6 N). CONCLUSIONS: The results indicate that the operation technique is feasible, and the stability of the EFF is sufficient for maintaining the reduction. The required pins can safely be inserted into the described areas with good reduction results. Using EFF offers a feasible alternative to the non-surgical treatment of CMF in LIC.

2.
Sci Rep ; 13(1): 3760, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882436

ABSTRACT

Neuroblastoma (NB) is a highly aggressive extracranial solid tumor in children. Due to its heterogeneity, NB remains a therapeutic challenge. Several oncogenic factors, including the Hippo effectors YAP/TAZ, are associated with NB tumorigenesis. Verteporfin (VPF) is an FDA-approved drug shown to directly inhibit YAP/TAZ activity. Our study aimed to investigate VPF's potential as a therapeutic agent in NB. We show that VPF selectively and efficiently impairs the viability of YAP/TAZ-expressing NB GI-ME-N and SK-N-AS cells, but not of non-malignant fibroblasts. To investigate whether VPF-mediated NB cell killing is YAP-dependent, we tested VPF potency in CRISPR-mediated YAP/TAZ knock-out GI-ME-N cells, and BE(2)-M17 NB cells (a MYCN-amplified, predominantly YAP-negative NB subtype). Our data shows that VPF-mediated NB cell killing is not dependent on YAP expression. Moreover, we determined that the formation of higher molecular weight (HMW) complexes is an early and shared VPF-induced cytotoxic mechanism in both YAP-positive and YAP-negative NB models. The accumulation of HMW complexes, involving STAT3, GM130 and COX IV proteins, impaired cell homeostasis and triggered cell stress and cell death mechanisms. Altogether, our study shows significant in vitro and in vivo VPF-induced suppression of NB growth, making VPF a potential therapeutic candidate against NB.


Subject(s)
Neuroblastoma , Child , Humans , Verteporfin/pharmacology , Aggression , Carcinogenesis , Homeostasis
3.
Cells ; 11(23)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36497012

ABSTRACT

Limbal stem cell deficiency (LSCD) is a complex, multifactorial disease affecting limbal epithelial progenitor cells (LEPC), which are essential for maintaining corneal stability and transparency. Human induced pluripotent stem cell-derived (hiPSC-) LEPC are a promising cell source for the treatment of LSCD. However, their similarity to native tissue-derived (T-) LEPC and their functional characterization has not been studied in detail. Here, we show that hiPSC-LEPC and T-LEPC have rather similar gene expression patterns, colony-forming ability, wound-healing capacity, and melanosome uptake. In addition, hiPSC-LEPC exhibited lower immunogenicity and reduced the proliferation of peripheral blood mononuclear cells compared with T-LEPC. Similarly, the hiPSC-LEPC secretome reduced the proliferation of vascular endothelial cells more than the T-LEPC secretome. Moreover, hiPSC-LEPC successfully repopulated decellularized human corneolimbal (DHC/L) scaffolds with multilayered epithelium, while basal deposition of fibrillary material was observed. These findings suggest that hiPSC-LEPC exhibited functional properties close to native LEPC and that hiPSC-LEPC-DHC/L scaffolds might be feasible for transplantation in patients suffering from LSCD in the future. Although hiPSC-LEPC-based stem cell therapy is promising, the current study also revealed new challenges, such as abnormal extracellular matrix deposition, that need to be overcome before hiPSC-LEPC-based stem cell therapies are viable.


Subject(s)
Epithelium, Corneal , Induced Pluripotent Stem Cells , Limbus Corneae , Humans , Epithelium, Corneal/metabolism , Endothelial Cells , Leukocytes, Mononuclear
4.
Stem Cell Rev Rep ; 18(8): 2952-2965, 2022 12.
Article in English | MEDLINE | ID: mdl-35727432

ABSTRACT

The neural crest gives rise to progeny as diverse as peripheral neurons, myelinating cells, cranial muscle, bone and cartilage tissues, and melanocytes. Neural crest derivation encompasses complex morphological change, including epithelial-to-mesenchymal transition (EMT) and migration to the eventual target locations throughout the body. Neural crest cultures derived from stem cells provide an attractive source for developmental studies in human model systems, of immediate biomedical relevance for neurocristopathies, neural cancer biology and regenerative medicine, if only appropriate markers for lineage and cell type definition and quality control criteria were available. Implementing a defined, scalable protocol to generate neural crest cells from embryonic stem cells, we identify stage-defining cluster-of-differentiation (CD) surface markers during human neural crest development in vitro. Acquisition of increasingly mesenchymal phenotype was characterized by absence of neuroepithelial stemness markers (CD15, CD133, CD49f) and by decrease of CD57 and CD24. Increased per-cell-expression of CD29, CD44 and CD73 correlated with established EMT markers as determined by immunofluorescence and immunoblot analysis. The further development towards migratory neural crest was associated with decreased CD24, CD49f (ITGA6) and CD57 (HNK1) versus an enhanced CD49d (ITGA4), CD49e (ITGA5) and CD51/CD61 (ITGAV/ITGB3) expression. Notably, a shift from CD57 to CD51/CD61 was identified as a sensitive surrogate surface indicator of EMT in neural crest in vitro development. The reported changes in glycan epitope and integrin surface expression may prove useful for elucidating neural crest stemness, EMT progression and malignancies.


Subject(s)
Embryonic Stem Cells , Neural Crest , Humans , Integrin alpha6/metabolism , Epitopes , Cell Differentiation , Biomarkers/metabolism
5.
Stem Cells ; 37(10): 1293-1306, 2019 10.
Article in English | MEDLINE | ID: mdl-31381839

ABSTRACT

Cell state-, developmental stage-, and lineage-specific combinatorial expression of cluster of differentiation (CD) molecules enables the identification of cellular subsets via multicolor flow cytometry. We describe an exhaustive characterization of neural cell types by surface antigens, exploiting human pluripotent stem cell-derived neural cell systems. Using multiwell screening approaches followed by detailed validation of expression patterns and dynamics, we exemplify a strategy for resolving cellular heterogeneity in stem cell paradigms. In addition to providing a catalog of surface antigens expressed in the neural lineage, we identified the transferrin receptor-1 (CD71) to be differentially expressed in neural stem cells and differentiated neurons. In this context, we describe a role for N-Myc proto-oncogene (MYCN) in maintaining CD71 expression in proliferating neural cells. We report that in vitro human stem cell-derived neurons lack CD71 surface expression and that the observed differential expression can be used to identify and enrich CD71- neuronal derivatives from heterogeneous cultures. Stem Cells 2019;37:1293-1306.


Subject(s)
Antigens, CD/metabolism , Antigens, Surface/metabolism , Biomarkers/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Transferrin/metabolism , Cell Differentiation , Flow Cytometry , Humans , Proto-Oncogene Mas
6.
Adv Mater ; 31(12): e1806727, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30687975

ABSTRACT

Interrogation and control of cellular fate and function using optogenetics is providing revolutionary insights into biology. Optogenetic control of cells is achieved by coupling genetically encoded photoreceptors to cellular effectors and enables unprecedented spatiotemporal control of signaling processes. Here, a fast and reversibly switchable photoreceptor is used to tune the mechanical properties of polymer materials in a fully reversible, wavelength-specific, and dose- and space-controlled manner. By integrating engineered cyanobacterial phytochrome 1 into a poly(ethylene glycol) matrix, hydrogel materials responsive to light in the cell-compatible red/far-red spectrum are synthesized. These materials are applied to study in human mesenchymal stem cells how different mechanosignaling pathways respond to changing mechanical environments and to control the migration of primary immune cells in 3D. This optogenetics-inspired matrix allows fundamental questions of how cells react to dynamic mechanical environments to be addressed. Further, remote control of such matrices can create new opportunities for tissue engineering or provide a basis for optically stimulated drug depots.

7.
Cell Rep ; 23(10): 2976-2988, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29874584

ABSTRACT

While mitochondrial dysfunction is emerging as key in Parkinson's disease (PD), a central question remains whether mitochondria are actual disease drivers and whether boosting mitochondrial biogenesis and function ameliorates pathology. We address these questions using patient-derived induced pluripotent stem cells and Drosophila models of GBA-related PD (GBA-PD), the most common PD genetic risk. Patient neurons display stress responses, mitochondrial demise, and changes in NAD+ metabolism. NAD+ precursors have been proposed to ameliorate age-related metabolic decline and disease. We report that increasing NAD+ via the NAD+ precursor nicotinamide riboside (NR) significantly ameliorates mitochondrial function in patient neurons. Human neurons require nicotinamide phosphoribosyltransferase (NAMPT) to maintain the NAD+ pool and utilize NRK1 to synthesize NAD+ from NAD+ precursors. Remarkably, NR prevents the age-related dopaminergic neuronal loss and motor decline in fly models of GBA-PD. Our findings suggest NR as a viable clinical avenue for neuroprotection in PD and other neurodegenerative diseases.


Subject(s)
Drosophila melanogaster/physiology , Induced Pluripotent Stem Cells/pathology , Mitochondria/pathology , NAD/metabolism , Neurons/metabolism , Neurons/pathology , Niacinamide/analogs & derivatives , Parkinson Disease/pathology , Animals , Autophagy , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endoplasmic Reticulum Stress , Glucosylceramidase/metabolism , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Dynamics , Motor Activity , Niacinamide/metabolism , Parkinson Disease/physiopathology , Pyridinium Compounds , Unfolded Protein Response
8.
Sci Rep ; 7(1): 13612, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051534

ABSTRACT

Neuroblastoma is the most common extra-cranial solid tumor in children. Its broad spectrum of clinical outcomes reflects the underlying inherent cellular heterogeneity. As current treatments often do not lead to tumor eradication, there is a need to better define therapy-resistant neuroblastoma and to identify new modulatory molecules. To this end, we performed the first comprehensive flow cytometric characterization of surface molecule expression in neuroblastoma cell lines. Exploiting an established clustering algorithm (SPADE) for unbiased visualization of cellular subsets, we conducted a multiwell screen for small molecule modulators of neuroblastoma phenotype. In addition to SH-SY5Y cells, the SH-EP, BE(2)-M17 and Kelly lines were included in follow-up analysis as in vitro models of neuroblastoma. A combinatorial detection of glycoprotein epitopes (CD15, CD24, CD44, CD57, TrkA) and the chemokine receptor CXCR4 (CD184) enabled the quantitative identification of SPADE-defined clusters differentially responding to small molecules. Exposure to bone morphogenetic protein (BMP)-4 was found to enhance a TrkAhigh/CD15-/CD184- neuroblastoma cellular subset, accompanied by a reduction in doublecortin-positive neuroblasts and of NMYC protein expression in SH-SY5Y cells. Beyond yielding novel marker candidates for studying neuroblastoma pathology, our approach may provide tools for improved pharmacological screens towards developing novel avenues of neuroblastoma diagnosis and treatment.


Subject(s)
Antigens, Surface/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Cluster Analysis , Humans , Lewis X Antigen/metabolism , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , RNA, Small Interfering/metabolism , Receptor, trkA/metabolism
9.
Neurobiol Dis ; 99: 133-144, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27993646

ABSTRACT

A cell's surface molecular signature enables its reciprocal interactions with the associated microenvironments in development, tissue homeostasis and pathological processes. The CD24 surface antigen (heat-stable antigen, nectadrin; small cell lung cancer antigen cluster-4) represents a prime example of a neural surface molecule that has long been known, but whose diverse molecular functions in intercellular communication we have only begun to unravel. Here, we briefly summarize the molecular fundamentals of CD24 structure and provide a comprehensive review of CD24 expression and functional studies in mammalian neural developmental systems and disease models (rodent, human). Striving for an integrated view of the intracellular signaling processes involved, we discuss the most pertinent routes of CD24-mediated signaling pathways and functional networks in neurobiology (neural migration, neurite extension, neurogenesis) and pathology (tumorigenesis, multiple sclerosis).


Subject(s)
CD24 Antigen/metabolism , Neurons/metabolism , Animals , Humans , Nervous System/growth & development , Nervous System/metabolism , Nervous System Diseases/metabolism
10.
Sci Rep ; 6: 23208, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26980066

ABSTRACT

The Hippo/YAP pathway serves as a major integrator of cell surface-mediated signals and regulates key processes during development and tumorigenesis. The neural crest is an embryonic tissue known to respond to multiple environmental cues in order to acquire appropriate cell fate and migration properties. Using multiple in vitro models of human neural development (pluripotent stem cell-derived neural stem cells; LUHMES, NTERA2 and SH-SY5Y cell lines), we investigated the role of Hippo/YAP signaling in neural differentiation and neural crest development. We report that the activity of YAP promotes an early neural crest phenotype and migration, and provide the first evidence for an interaction between Hippo/YAP and retinoic acid signaling in this system.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neural Crest/cytology , Neural Stem Cells/physiology , Neurogenesis , Phosphoproteins/physiology , Antigens, CD/metabolism , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Humans , Signal Transduction , Transcription Factors , Tretinoin/pharmacology , YAP-Signaling Proteins
11.
J Vis Exp ; (94)2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25549236

ABSTRACT

Flow cytometry has been extensively used to define cell populations in immunology, hematology and oncology. Here, we provide a detailed description of protocols for flow cytometric analysis of the cluster of differentiation (CD) surface antigens and intracellular antigens in neural cell types. Our step-by-step description of the methodological procedures include: the harvesting of neural in vitro cultures, an optional carboxyfluorescein succinimidyl ester (CFSE)-labeling step, followed by surface antigen staining with conjugated CD antibodies (e.g., CD24, CD54), and subsequent intracellar antigen detection via primary/secondary antibodies or fluorescently labeled Fab fragments (Zenon labeling). The video demonstrates the most critical steps. Moreover, principles of experimental planning, the inclusion of critical controls, and fundamentals of flow cytometric analysis (identification of target population and exclusion of debris; gating strategy; compensation for spectral overlap) are briefly explained in order to enable neurobiologists with limited prior knowledge or specific training in flow cytometry to assess its utility and to better exploit this powerful methodology.


Subject(s)
Antigens, CD/analysis , Flow Cytometry/methods , Neural Stem Cells/immunology , Antigens, Surface/analysis , Fluoresceins/chemistry , Humans , Immunoconjugates/chemistry , Immunoglobulin Variable Region/chemistry , Neural Stem Cells/chemistry , Neural Stem Cells/cytology , Staining and Labeling/methods
12.
Hum Mol Genet ; 23(23): 6177-90, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24990151

ABSTRACT

Simultaneous generation of neural cells and that of the nutrient-supplying vasculature during brain development is called neurovascular coupling. We report on a transgenic mouse with impaired transforming growth factor ß (TGFß)-signalling in forebrain-derived neural cells using a Foxg1-cre knock-in to drive the conditional knock-out of the Tgfbr2. Although the expression of FOXG1 is assigned to neural progenitors and neurons of the telencephalon, Foxg1(cre/+);Tgfbr2(flox/flox) (Tgfbr2-cKO) mutants displayed intracerebral haemorrhage. Blood vessels exhibited an atypical, clustered appearance were less in number and displayed reduced branching. Vascular endothelial growth factor (VEGF) A, insulin-like growth factor (IGF) 1, IGF2, TGFß, inhibitor of DNA binding (ID) 1, thrombospondin (THBS) 2, and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) 1 were altered in either expression levels or tissue distribution. Accordingly, human umbilical vein endothelial cells (HUVEC) displayed branching defects after stimulation with conditioned medium (CM) that was derived from primary neural cultures of the ventral and dorsal telencephalon of Tgfbr2-cKO. Supplementing CM of Tgfbr2-cKO with VEGFA rescued these defects, but application of TGFß aggravated them. HUVEC showed reduced migration towards CM of mutants compared with controls. Supplementing the CM with growth factors VEGFA, fibroblast growth factor (FGF) 2 and IGF1 partially restored HUVEC migration. In contrast, TGFß supplementation further impaired migration of HUVEC. We observed differences along the dorso-ventral axis of the telencephalon with regard to the impact of these factors on the phenotype. Together these data establish a TGFBR2-dependent molecular crosstalk between neural and endothelial cells during brain vessel development. These findings will be useful to further elucidate neurovascular interaction in general and to understand pathologies of the blood vessel system such as intracerebral haemorrhages, hereditary haemorrhagic telangiectasia, Alzheimers disease, cerebral amyloid angiopathy or tumour biology.


Subject(s)
Brain/metabolism , Neovascularization, Physiologic , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/pathology , Cell Movement , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Culture Media, Conditioned , Fibroblast Growth Factor 2/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurons/pathology , Pericytes/metabolism , Pericytes/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Secretory Pathway , Telencephalon/blood supply , Telencephalon/metabolism , Telencephalon/pathology , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Nat Commun ; 5: 4028, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24905578

ABSTRACT

Mutations in the acid ß-glucocerebrosidase (GBA1) gene, responsible for the lysosomal storage disorder Gaucher's disease (GD), are the strongest genetic risk factor for Parkinson's disease (PD) known to date. Here we generate induced pluripotent stem cells from subjects with GD and PD harbouring GBA1 mutations, and differentiate them into midbrain dopaminergic neurons followed by enrichment using fluorescence-activated cell sorting. Neurons show a reduction in glucocerebrosidase activity and protein levels, increase in glucosylceramide and α-synuclein levels as well as autophagic and lysosomal defects. Quantitative proteomic profiling reveals an increase of the neuronal calcium-binding protein 2 (NECAB2) in diseased neurons. Mutant neurons show a dysregulation of calcium homeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium. Importantly, correction of the mutations rescues such pathological phenotypes. These findings provide evidence for a link between GBA1 mutations and complex changes in the autophagic/lysosomal system and intracellular calcium homeostasis, which underlie vulnerability to neurodegeneration.


Subject(s)
Autophagy , Calcium/metabolism , Glucosylceramidase/genetics , Homeostasis , Induced Pluripotent Stem Cells/pathology , Neurons/pathology , Parkinson Disease/pathology , Cell Differentiation , Glycoside Hydrolases/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Neurons/enzymology , Parkinson Disease/immunology , Parkinson Disease/metabolism
14.
Mol Cell Ther ; 2: 2, 2014.
Article in English | MEDLINE | ID: mdl-26056571

ABSTRACT

For a range of nervous system disorders current treatment options remain limited. Focusing on Parkinson's disease as a neurodegenerative entity that affects an increasing quantity of people in our aging societies, we briefly discuss remaining challenges and opportunities that neural stem cell therapy might be able to offer. Providing a snapshot of neural transplantation paradigms, we contemplate possible imminent translational scenarios and discuss critical requirements to be considered before clinical implementation.

15.
Methods Mol Biol ; 1037: 117-44, 2013.
Article in English | MEDLINE | ID: mdl-24029933

ABSTRACT

The nervous system is characterized by its complex network of highly specialized cells that enable us to perceive stimuli from the outside world and react accordingly. The computational integration enabled by these networks remains to be elucidated, but appropriate sensory input, processing, and motor control are certainly essential for survival. Consequently, loss of nervous tissue due to injury or disease represents a considerable biomedical challenge. Stem cell research offers the promise to provide cells for nervous system repair to replace lost and damaged neural tissue and alleviate disease. We provide a protocol-based chapter on fundamental principles and procedures of pluripotent stem cell (PSC) differentiation and neural transplantation. Rather than detailed methodological step-by-step descriptions of these procedures, we provide an overview and highlight the most critical aspects and key steps of PSC neural induction, subtype specification in different in vitro systems, as well as neural cell transplantation to the central nervous system. We conclude with a summary of suitable readout methods including in vitro phenotypic analysis, histology, and functional analysis in vivo.


Subject(s)
Nerve Regeneration , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Central Nervous System/metabolism , Disease Models, Animal , Humans , Mice , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Neurons/cytology , Neurons/metabolism , Phenotype , Rats , Stem Cell Transplantation/methods
16.
PLoS One ; 8(6): e68519, 2013.
Article in English | MEDLINE | ID: mdl-23826393

ABSTRACT

Surface molecule profiles undergo dynamic changes in physiology and pathology, serve as markers of cellular state and phenotype and can be exploited for cell selection strategies and diagnostics. The isolation of well-defined cell subsets is needed for in vivo and in vitro applications in stem cell biology. In this technical report, we present an approach for defining a subset of interest in a mixed cell population by flow cytometric detection of intracellular antigens. We have developed a fully validated protocol that enables the co-detection of cluster of differentiation (CD) surface antigens on fixed, permeabilized neural cell populations defined by intracellular staining. Determining the degree of co-expression of surface marker candidates with intracellular target population markers (nestin, MAP2, doublecortin, TUJ1) on neuroblastoma cell lines (SH-SY5Y, BE(2)-M17) yielded a combinatorial CD49f(-)/CD200(high) surface marker panel. Its application in fluorescence-activated cell sorting (FACS) generated enriched neuronal cultures from differentiated cell suspensions derived from human induced pluripotent stem cells. Our data underlines the feasibility of using the described co-labeling protocol and co-expression analysis for quantitative assays in mammalian neurobiology and for screening approaches to identify much needed surface markers in stem cell biology.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation , Cell Membrane/metabolism , Flow Cytometry/methods , Intracellular Space/metabolism , Neurons/cytology , Neurons/metabolism , Biomarkers/metabolism , Cell Line , Cell Separation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Reproducibility of Results
17.
Cell Transplant ; 22(7): 1281-93, 2013.
Article in English | MEDLINE | ID: mdl-22963760

ABSTRACT

Cell replacement therapy by intracerebral transplantation of fetal dopaminergic neurons has become a promising therapeutic option for patients suffering from Parkinson's disease during the last decades. However, limited availability of human fetal tissue as well as ethical issues, lack of alternative nonfetal donor cells, and the absence of standardized transplantation protocols have prevented neurorestorative therapies from becoming a routine procedure in patients suffering from neurodegenerative diseases. Improvement of graft survival, surgery techniques, and identification of the optimal target area are imperative for further optimization of this novel treatment. In the present study, human primary fetal ventral mesencephalon-derived tissue from 7- to 9-week-old human fetuses was transplanted into 6-hydroxydopamine-lesioned adult Sprague-Dawley rats. Graft survival, fiber outgrowth, and drug-induced rotational behavior up to 14 weeks posttransplantation were compared between different intrastriatal transplantation techniques (full single cell suspension vs. partial tissue pieces suspension injected by glass capillary or metal cannula) and the intranigral glass capillary injection of a full (single cell) suspension. The results demonstrate a higher survival rate of dopamine neurons, a greater reduction in amphetamine-induced rotations (overcompensation), and more extensive fiber outgrowth for the intrastriatally transplanted partial (tissue pieces) suspension compared to all other groups. Apomorphine-induced rotational bias was significantly reduced in all groups including the intranigral group. The data confirm that human ventral mesencephalon-derived cells serve as a viable cell source, survive in a xenografting paradigm, and functionally integrate into the host tissue. In contrast to rat donor cells, keeping the original (fetal) neuronal network by preparing only a partial suspension containing tissue pieces seems to be beneficial for human cells, although a metal cannula that causes greater tissue trauma to the host is required for injection. In addition, homotopic intranigral grafts may represent a complimentary grafting approach to the "classical" ectopic intrastriatal target site in PD.


Subject(s)
Mesencephalon/cytology , Parkinson Disease/therapy , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Disease Models, Animal , Embryo, Mammalian/cytology , Female , Graft Survival , Humans , Mesencephalon/transplantation , Neurons/metabolism , Neurons/pathology , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Transplantation, Heterologous , Tyrosine 3-Monooxygenase/metabolism
18.
Stem Cell Rev Rep ; 8(3): 813-29, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22628111

ABSTRACT

Pluripotent stem cells offer an abundant and malleable source for the generation of differentiated cells for transplantation as well as for in vitro screens. Patterning and differentiation protocols have been developed to generate neural progeny from human embryonic or induced pluripotent stem cells. However, continued refinement is required to enhance efficiency and to prevent the generation of unwanted cell types. We summarize and interpret insights gained from studies of embryonic neuroepithelium. A multitude of factors including soluble molecules, interactions with the extracellular matrix and neighboring cells cooperate to control neural stem cell self-renewal versus differentiation. Applying these findings and concepts to human stem cell systems in vitro may yield more appropriately patterned cell types for biomedical applications.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Neural Stem Cells/physiology , Pluripotent Stem Cells/physiology , Stem Cell Niche , Animals , Cell Communication , Cell Polarity , Embryonic Stem Cells/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Extracellular Matrix Proteins/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/metabolism , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction
19.
Cell ; 144(5): 782-95, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376238

ABSTRACT

During development and regeneration, proliferation of tissue-specific stem cells is tightly controlled to produce organs of a predetermined size. The molecular determinants of this process remain poorly understood. Here, we investigate the function of Yap1, the transcriptional effector of the Hippo signaling pathway, in skin biology. Using gain- and loss-of-function studies, we show that Yap1 is a critical modulator of epidermal stem cell proliferation and tissue expansion. Yap1 mediates this effect through interaction with TEAD transcription factors. Additionally, our studies reveal that α-catenin, a molecule previously implicated in tumor suppression and cell density sensing in the skin, is an upstream negative regulator of Yap1. α-catenin controls Yap1 activity and phosphorylation by modulating its interaction with 14-3-3 and the PP2A phosphatase. Together, these data identify Yap1 as a determinant of the proliferative capacity of epidermal stem cells and as an important effector of a "crowd control" molecular circuitry in mammalian skin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Epidermal Cells , Phosphoproteins/metabolism , alpha Catenin/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Cycle Proteins , Cell Line , Epidermis/metabolism , Mice , YAP-Signaling Proteins
20.
Blood ; 115(20): 4039-42, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20233975

ABSTRACT

Induced pluripotent stem cells (iPSCs) can be generated from various differentiated cell types by the expression of a set of defined transcription factors. So far, iPSCs have been generated from primary cells, but it is unclear whether human cancer cell lines can be reprogrammed. Here we describe the generation and characterization of iPSCs derived from human chronic myeloid leukemia cells. We show that, despite the presence of oncogenic mutations, these cells acquired pluripotency by the expression of 4 transcription factors and underwent differentiation into cell types derived of all 3 germ layers during teratoma formation. Interestingly, although the parental cell line was strictly dependent on continuous signaling of the BCR-ABL oncogene, also termed oncogene addiction, reprogrammed cells lost this dependency and became resistant to the BCR-ABL inhibitor imatinib. This finding indicates that the therapeutic agent imatinib targets cells in a specific epigenetic differentiated cell state, and this may contribute to its inability to fully eradicate disease in chronic myeloid leukemia patients.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Benzamides , Cells, Cultured , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...