Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(6): e0199996, 2018.
Article in English | MEDLINE | ID: mdl-29953534

ABSTRACT

Here we provide new technology for generating human peptidergic nociceptive sensory neurons in a straightforward and efficient way. The cellular source, human epidermal neural crest stem cells (hEPI-NCSC), consists of multipotent somatic stem cells that reside in the bulge of hair follicles. hEPI-NCSC and primary sensory neurons have a common origin, the embryonic neural crest. For directed differentiation, hEPI-NCSC were exposed to pertinent growth factors and small molecules in order to modulate master signalling networks involved in differentiation of neural crest cells into postmitotic peptidergic sensory neurons during embryonic development. The neuronal populations were homogenous in regard to antibody marker expression. Cells were immunoreactive for essential master regulatory genes, including NGN1/2, SOX10, and BRN3a among others, and for the pain-mediating genes substance P (SP), calcitonin gene related protein (CGRP) and the TRPV1 channel. Approximately 30% of total cells responded to capsaicin, indicating that they expressed an active TRPV1 channel. In summary, hEPI-NCSC are a biologically relevant and easily available source of somatic stem cells for generating human peptidergic nociceptive neurons without the need for genetic manipulation and cell purification. As no analgesics exist that specifically target TRPV1, a ready supply of high-quality human peptidergic nociceptive sensory neurons could open the way for new approaches, in a biologically relevant cellular context, to drug discovery and patient-specific disease modelling that is aimed at pain control, and as such is highly desirable.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Multipotent Stem Cells/metabolism , Neural Crest/metabolism , Nociceptors/metabolism , Signal Transduction , Humans , Multipotent Stem Cells/cytology , Neural Crest/cytology , Nociceptors/cytology
2.
Development ; 142(18): 3188-97, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26251357

ABSTRACT

We show that highly pure populations of human Schwann cells can be derived rapidly and in a straightforward way, without the need for genetic manipulation, from human epidermal neural crest stem cells [hEPI-NCSC(s)] present in the bulge of hair follicles. These human Schwann cells promise to be a useful tool for cell-based therapies, disease modelling and drug discovery. Schwann cells are glia that support axons of peripheral nerves and are direct descendants of the embryonic neural crest. Peripheral nerves are damaged in various conditions, including through trauma or tumour-related surgery, and Schwann cells are required for their repair and regeneration. Schwann cells also promise to be useful for treating spinal cord injuries. Ex vivo expansion of hEPI-NCSC isolated from hair bulge explants, manipulating the WNT, sonic hedgehog and TGFß signalling pathways, and exposure of the cells to pertinent growth factors led to the expression of the Schwann cell markers SOX10, KROX20 (EGR2), p75NTR (NGFR), MBP and S100B by day 4 in virtually all cells, and maturation was completed by 2 weeks of differentiation. Gene expression profiling demonstrated expression of transcripts for neurotrophic and angiogenic factors, as well as JUN, all of which are essential for nerve regeneration. Co-culture of hEPI-NCSC-derived human Schwann cells with rodent dorsal root ganglia showed interaction of the Schwann cells with axons, providing evidence of Schwann cell functionality. We conclude that hEPI-NCSCs are a biologically relevant source for generating large and highly pure populations of human Schwann cells.


Subject(s)
Adult Stem Cells/physiology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Lineage/physiology , Neural Crest/physiology , Schwann Cells/cytology , Gene Expression Profiling , Humans , Immunohistochemistry , Real-Time Polymerase Chain Reaction , Schwann Cells/physiology
3.
Stem Cells Transl Med ; 4(10): 1173-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26273065

ABSTRACT

UNLABELLED: This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE: It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.


Subject(s)
Neural Crest/transplantation , Neural Stem Cells/transplantation , Spinal Cord/cytology , Stem Cell Transplantation/methods , Animals , Behavior, Animal , Cell Survival , Dogs , Epidermal Cells , Feasibility Studies , Gait , Injections, Spinal , Magnetic Resonance Imaging , Mice , Mice, Knockout , Neurogenesis , Pilot Projects , Stem Cell Transplantation/adverse effects , Teratoma , Walking
4.
Stem Cell Rev Rep ; 11(1): 180-93, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25173879

ABSTRACT

The use of cell transplantation for spinal cord injury is a rapidly evolving field in regenerative medicine. Numerous animal models are currently being used. However, translation to human patients is still a challenging step. Dogs are of increasing importance as a translational model for human disease since there is a greater awareness of the need to increase the quality of preclinical data. The use of dogs ultimately brings benefit to both human and veterinary medicine. In this review we analyze experimental and clinical studies using cell transplantation for canine spinal cord injury. Overall, in experimental studies, transplantation groups showed improvement over control groups. Improvements were measured at the functional, electrophysiological, histological, RNA and protein levels. Most clinical studies support beneficial effects of cell transplantation despite the fact that methodological limitations preclude definitive conclusions. However, the mechanisms of action and underlying the behavior of transplanted cells in the injured spinal cord remain unclear. Overall, we conclude here that stem cell interventions are a promising avenue for the treatment of spinal cord injury. Canines are a promising model that may help bridge the gap between translational research and human clinical trials.


Subject(s)
Disease Models, Animal , Regenerative Medicine/methods , Spinal Cord Injuries/therapy , Stem Cell Transplantation/methods , Animals , Dogs , Humans , Recovery of Function , Regenerative Medicine/trends , Spinal Cord Injuries/physiopathology , Transplantation, Heterologous , Transplantation, Homologous
5.
Birth Defects Res C Embryo Today ; 102(3): 221-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25228472

ABSTRACT

In this review article I explore the suitability of human epidermal neural crest stem cells (hEPI-NCSC) for translational medicine. hEPI-NCSC are multipotent somatic stem cells that are derived from the embryonic neural crest. hEPI-NCSC are located in the bulge of hair follicles where they persist postnatally and into adulthood. Because of their location in the hairy skin and their migratory behavior, hEPI-NCSC can be easily isolated as a highly pure population of stem cells without the need for purification. Furthermore they can be expanded ex vivo into millions of stem cells, they do not form tumors in vivo, and they can undergo directed differentiation into crest and noncrest-derived cell types of clinical relevance. Taken together, these characteristics make hEPI-NCSC attractive candidates for cell-based therapies, drug discovery, and disease modeling.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Drug Discovery/methods , Neural Crest/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Disease Models, Animal , Humans , Spinal Cord Injuries/therapy
6.
Stem Cell Rev Rep ; 10(2): 316-26, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24399192

ABSTRACT

Here we provide a protocol for the directed differentiation of hEPI-NCSC into midbrain dopaminergic neurons, which degenerate in Parkinson's disease. hEPI-NCSC are neural crest-derived multipotent stem cells that persist into adulthood in the bulge of hair follicles. The experimental design is distinctly different from conventional protocols for embryonic stem cells and induced pluripotent stem (iPS) cells. It includes pre-differentiation of the multipotent hEPI-NCSC into neural stem cell-like cells, followed by ventralizing, patterning, continued exposure to the TGFß receptor inhibitor, SB431542, and at later stages of differentiation the presence of the WNT inhibitor, IWP-4. All cells expressed A9 midbrain dopaminergic neuron progenitor markers with gene expression levels comparable to those in normal human substantia nigra. The current study shows for the first time that virtually homogeneous populations of dopaminergic neurons can be derived ex vivo from somatic stem cells without the need for purification, with useful timeliness and high efficacy. This novel development is an important first step towards the establishment of fully functional dopaminergic neurons from an ontologically relevant stem cell type, hEPI-NCSC.


Subject(s)
Cell Differentiation , Dopaminergic Neurons/metabolism , Neural Stem Cells/physiology , Cell Culture Techniques , Cells, Cultured , Dopaminergic Neurons/cytology , Epidermal Cells , Humans , Neural Crest/cytology , Substantia Nigra/cytology
7.
Stem Cells Transl Med ; 3(3): 334-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24443004

ABSTRACT

The discovery of multipotent neural crest-derived stem cells, named epidermal neural crest stem cells (EPI-NCSC), that persist postnatally in an easy-to-access location-the bulge of hair follicles-opens a spectrum of novel opportunities for patient-specific therapies. We present a detailed characterization of canine EPI-NCSC (cEPI-NCSC) from multiple dog breeds and protocols for their isolation and ex vivo expansion. Furthermore, we provide novel tools for research in canines, which currently are still scarce. In analogy to human and mouse EPI-NCSC, the neural crest origin of cEPI-NCSC is shown by their expression of the neural crest stem cell molecular signature and other neural crest-characteristic genes. Similar to human EPI-NCSC, cEPI-NCSC also expressed pluripotency genes. We demonstrated that cEPI-NCSC can generate all major neural crest derivatives. In vitro clonal analyses established multipotency and self-renewal ability of cEPI-NCSC, establishing cEPI-NCSC as multipotent somatic stem cells. A critical analysis of the literature on canine spinal cord injury (SCI) showed the need for novel treatments and suggested that cEPI-NCSC represent viable candidates for cell-based therapies in dog SCI, particularly for chondrodystrophic dogs. This notion is supported by the close ontological relationship between neural crest stem cells and spinal cord stem cells. Thus, cEPI-NCSC promise to offer not only a potential treatment for canines but also an attractive and realistic large animal model for human SCI. Taken together, we provide the groundwork for the development of a novel cell-based therapy for a condition with extremely poor prognosis and no available effective treatment.


Subject(s)
Hair Follicle/cytology , Multipotent Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Proliferation , Cell Separation , Cell- and Tissue-Based Therapy , Disease Models, Animal , Dogs , Female , Gene Expression , Hair Follicle/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/transplantation , Neural Crest/metabolism , Neural Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy
8.
Stem Cell Rev Rep ; 10(1): 60-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23955574

ABSTRACT

We show that neural crest stem cells affect mouse hair follicle development. During embryogenesis hair follicle induction is regulated by complex reciprocal and functionally redundant signals between epidermis and dermis, which remain to be fully understood. Canonical Wnt signalling is a hallmark of neural crest cells and also a prerequisite for hair follicle induction prior to hair placode formation in the epidermis. As neural crest stem cells invade the epidermis during early embryonic development we aimed at determining whether neural crest cells affect hair follicle development. To attenuate, but not silence, canonical Wnt signalling specifically in neural crest cells, we analyzed Wnt1-cre(+/-)::Pygo2(-/-) mice in which the ß-catenin co-activator gene, Pygopus 2 (Pygo2), is deleted specifically in neural crest cells. Both, hair density and hair thickness were reduced in mutant mice. Furthermore, hair development was delayed and the relative ratio of hair types was affected. There was a decrease in zig-zag hairs and an increase in awl hairs. Mouse neural crest stem cells expressed ectodysplasin, an essential effector in the formation of zig-zag hair. Taken together, our data support the novel notion that neural crest cells are involved in the earliest stages of hair follicle development.


Subject(s)
Gene Deletion , Hair Follicle/growth & development , Hair Follicle/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neural Crest/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Hair Follicle/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Crest/metabolism
9.
Stem Cell Res Ther ; 2(3): 29, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21722345

ABSTRACT

Wong and Reiter have explored the possibility that hair follicle stem cells can give rise to basal cell carcinoma (BCC). They expressed in mice an inducible human BCC-derived oncogenic allele of Smoothened, SmoM2, under the control of either the cytokeratin 14 (K14) or cytokeratin 15 (K15) promoter. Smoothened encodes a G-protein-coupled receptor protein in the hedgehog pathway, the misregulation of which is implicated in BCC and other human cancers. Chronic injury is thought to be a contributing factor. The authors used K14 as a marker for stem cells in the basal layer of the epidermis and K15 as a marker for epidermal stem cells in the bulge of hair follicles. Upon activation, K14 construct-bearing mice readily formed BCC-like tumours, whereas this was not the case in K15:SmoM2-carrying mice. Upon wounding the epidermis, however, there was widespread BCC-like tumour formation in the skin of K15:SmoM2 mice. The authors conclude that wounding recruited bulge epidermal stem cells to the surface, allowing the cells to escape quiescence in the stem cell niche and to arrive in an environment where the hedgehog pathway becomes activated and therefore tumorigenesis is elicited. While this is a provocative result and the authors' conclusion may well be correct, there are alternative explanations.

10.
J Pathol ; 224(3): 309-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21598248

ABSTRACT

Patients carrying heterozygous germline truncating mutations in the CYLD gene develop multiple primary hair follicle-related tumours. A highly patterned tumour, termed cylindroma, and a highly disorganized tumour, termed spiradenoma, may both develop in the same patient. Furthermore, histological features of both tumour types have been described within the same tumour specimen. We used three-dimensional computer-aided reconstruction of these tumours to demonstrate contiguous growth of cylindromas into spiradenomas, thus suggesting a transition between the two tumour types. To explore factors that may influence cutaneous tumour patterning, genome-wide transcriptomic analysis of 32 CYLD-defective tumours was performed. Overexpression of the Wnt/ß-catenin signalling pathway was observed relative to normal perilesional tissue. Morphometric analysis was used to investigate the relationship between Wnt pathway-related gene expression and tumour organization. This revealed an association between reduced Dickkopf 2 (DKK2-a negative regulator of the Wnt/ß-catenin signalling pathway) expression and loss of tumour patterning. Reduced DKK2 expression was associated with methylation of the DKK2 gene promoter in the majority of tumour samples assayed. RNA interference-mediated silencing of DKK2 expression in cylindroma primary cell cultures caused an increase in colony formation, cell viability, and anchorage-independent growth. Using these data, we propose a model where epigenetic programming may influence tumour patterning in patients with CYLD mutations.


Subject(s)
Adenoma, Sweat Gland/metabolism , Carcinoma, Adenoid Cystic/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Skin Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Adenoma, Sweat Gland/genetics , Adenoma, Sweat Gland/pathology , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/pathology , Cell Proliferation , DNA Methylation , DNA, Neoplasm/genetics , Deubiquitinating Enzyme CYLD , Disease Progression , Gene Knockdown Techniques , Genome-Wide Association Study/methods , Germ-Line Mutation , Humans , Imaging, Three-Dimensional , Intercellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , Promoter Regions, Genetic , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sweat Gland Neoplasms/genetics , Sweat Gland Neoplasms/metabolism , Sweat Gland Neoplasms/pathology , Tumor Cells, Cultured , Wnt Proteins/metabolism , beta Catenin/metabolism
11.
Stem Cell Rev Rep ; 7(4): 799-814, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21455606

ABSTRACT

Here we describe the isolation, characterisation and ex-vivo expansion of human epidermal neural crest stem cells (hEPI-NCSC) and we provide protocols for their directed differentiation into osteocytes and melanocytes. hEPI-NCSC are neural crest-derived multipotent stem cells that persist into adulthood in the bulge of hair follicles. Multipotency and self-renewal were determined by in vitro clonal analyses. hEPI-NCSC generate all major neural crest derivatives, including bone/cartilage cells, neurons, Schwann cells, myofibroblasts and melanocytes. Furthermore, hEPI-NCSC express additional neural crest stem cell markers and global stem cell genes. To variable degrees and in a donor-dependent manner, hEPI-NCSC express the six essential pluripotency genes C-MYC, KLF4, SOX2, LIN28, OCT-4/POU5F1 and NANOG. hEPI-NCSC can be expanded ex vivo into millions of stem cells that remain mulitpotent and continue to express stem cell genes. The novelty of hEPI-NCSC lies in the combination of their highly desirable traits. hEPI-NCSC are embryonic remnants in a postnatal location, the bulge of hair follicles. Therefore they are readily accessible in the hairy skin by minimal invasive procedure. hEPI-NCSC are multipotent somatic stem cells that can be isolated reproducibly and with high yield. By taking advantage of their migratory ability, hEPI-NCSC can be isolated as a highly pure population of stem cells. hEPI-NCSC can undergo robust ex vivo expansion and directed differentiation. As somatic stem cells, hEPI-NCSC are conducive to autologous transplantation, which avoids graft rejection. Together, these traits make hEPI-NCSC novel and attractive candidates for future cell-based therapies and regenerative medicine.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Epidermal Cells , Melanocytes/cytology , Neural Crest/cytology , Osteocytes/cytology , Biomarkers/metabolism , Cell Culture Techniques , Cell Movement , Cells, Cultured , Cryopreservation , Gene Expression Regulation , Humans , Immunohistochemistry , Kruppel-Like Factor 4 , Schwann Cells/cytology
12.
Brain Res Bull ; 83(5): 189-93, 2010 Oct 30.
Article in English | MEDLINE | ID: mdl-20637266

ABSTRACT

Epidermal neural crest stem cell (EPI-NCSC) grafts cause a significant improvement in sensory connectivity and touch perception in the contused mouse spinal cord. EPI-NCSC are derived from the embryonic neural crest but reside in a postnatal location, the bulge of hair follicles. Both mouse and human EPI-NCSC are multipotent adult stem cells capable of generating all major neural crest derivatives. EPI-NCSC of mouse and human origin express the neural crest stem cell molecular signature, genes that were initially used to create induced pluripotent stem (iPS) cells, and other neural crest and global stem cell genes. Due to their origin in the neural folds and because they share a higher order stem cell, neural crest cells, and thus EPI-NCSC, are closely related to neural tube stem cells. This close ontological relationship with the spinal cord makes EPI-NCSC attractive candidates for cell-based therapy in spinal cord injury. In two different contusion models of spinal cord injury, we have shown that EPI-NCSC integrate into the murine spinal cord tissue and that subsets differentiate into GABAergic neurons and myelinating oligodendrocytes. Intraspinal EPI-NCSC do not form tumours. In the presence of EPI-NCSC grafts, but not in control animals, there is a 24% improvement of sensory connectivity and a substantial improvement in touch perception. Unilateral transplants leading to bilateral functional improvements suggest that underlying mechanisms include diffusible molecules. EPI-NCSC indeed express genes that encode neurotrophins, other trophic factors, angiogenic factors and metalloproteases. Intraspinal EPI-NCSC thus have multiple effects in the contused spinal cord, the sum of which can explain the observed functional improvements.


Subject(s)
Epidermis , Models, Animal , Multipotent Stem Cells/physiology , Neural Crest/cytology , Spinal Cord Injuries , Animals , Epidermal Cells , Epidermis/embryology , Humans , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
13.
Stem Cell Rev Rep ; 6(2): 186-98, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20414748

ABSTRACT

Here we show that epidermal neural crest stem cell (EPI-NCSC) transplants in the contused spinal cord caused a 24% improvement in sensory connectivity and a substantial recovery of touch perception. Furthermore we present a novel method for the ex vivo expansion of EPI-NCSC into millions of stem cells that takes advantage of the migratory ability of neural crest stem cells and is based on a new culture medium and the use of microcarriers. Functional improvement was shown by two independent methods, spinal somatosensory evoked potentials (SpSEP) and the Semmes-Weinstein touch test. Subsets of transplanted cells differentiated into myelinating oligodendrocytes. Unilateral injections of EPI-NCSC into the lesion of midline contused mouse spinal cords elicited bilateral improvements. Intraspinal EPI-NCSC did not migrate laterally in the spinal cord or invade the spinal roots and dorsal root ganglia, thus implicating diffusible factors. EPI-NCSC expressed neurotrophic factors, angiogenic factors, and metalloproteases. The strength of EPI-NCSC thus is that they can exert a combination of pertinent functions in the contused spinal cord, including cell replacement, neuroprotection, angiogenesis and modulation of scar formation. EPI-NCSC are uniquely qualified for cell-based therapy in spinal cord injury, as neural crest cells and neural tube stem cells share a higher order stem cell and are thus ontologically closely related.


Subject(s)
Spinal Cord Injuries/therapy , Animals , Epidermal Cells , Evoked Potentials, Somatosensory/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron, Transmission , Models, Biological , Neural Crest/cytology , Polymerase Chain Reaction , Spinal Cord/pathology , Spinal Cord/ultrastructure , Stem Cell Transplantation , Stem Cells/cytology
14.
Biomed Sci Instrum ; 45: 167-72, 2009.
Article in English | MEDLINE | ID: mdl-19369758

ABSTRACT

Diffusion weighted magnetic resonance imaging (DWI) is a powerful tool for evaluation of microstructural anomalies in numerous central nervous system pathologies. Diffusion tensor imaging (DTI) allows for the magnitude and direction of water self diffusion to be estimated by sampling the apparent diffusion coefficient (ADC) in various directions. Clinical DWI and DTI performed at a single level of diffusion weighting, however, does not allow for multiple diffusion compartments to be elicited. Furthermore, assumptions made regarding the precise number of diffusion compartments intrinsic to the tissue of interest have resulted in a lack of consensus between investigations. To overcome these challenges, a stretched-exponential model of diffusion was applied to examine the diffusion coefficient and "heterogeneity index" within highly compartmentalized brain tumors. The purpose of the current study is to expand on the stretched-exponential model of diffusion to include directionality of both diffusion heterogeneity and apparent diffusion coefficient. This study develops the mathematics of this new technique along with an initial application in quantifying spinal cord regeneration following acute injection of epidermal neural crest stem cell (EPI-NCSC) grafts.

15.
BMC Genomics ; 10: 151, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19356247

ABSTRACT

BACKGROUND: We have identified a differential gene expression profile in neural crest stem cells that is due to deletion of the norepinephrine transporter (NET) gene. NET is the target of psychotropic substances, such as tricyclic antidepressants and the drug of abuse, cocaine. NET mutations have been implicated in depression, anxiety, orthostatic intolerance and attention deficit hyperactivity disorder (ADHD). NET function in adult noradrenergic neurons of the peripheral and central nervous systems is to internalize norepinephrine from the synaptic cleft. By contrast, during embryogenesis norepinephrine (NE) transport promotes differentiation of neural crest stem cells and locus ceruleus progenitors into noradrenergic neurons, whereas NET inhibitors block noradrenergic differentiation. While the structure of NET und the regulation of NET function are well described, little is known about downstream target genes of norepinephrine (NE) transport. RESULTS: We have prepared gene expression profiles of in vitro differentiating wild type and norepinephrine transporter-deficient (NETKO) mouse neural crest cells using long serial analysis of gene expression (LongSAGE). Comparison analyses have identified a number of important differentially expressed genes, including genes relevant to neural crest formation, noradrenergic neuron differentiation and the phenotype of NETKO mice. Examples of differentially expressed genes that affect noradrenergic cell differentiation include genes in the bone morphogenetic protein (BMP) signaling pathway, the Phox2b binding partner Tlx2, the ubiquitin ligase Praja2, and the inhibitor of Notch signaling, Numbl. Differentially expressed genes that are likely to contribute to the NETKO phenotype include dopamine-beta-hydroxylase (Dbh), tyrosine hydroxylase (Th), the peptide transmitter 'cocaine and amphetamine regulated transcript' (Cart), and the serotonin receptor subunit Htr3a. Real-time PCR confirmed differential expression of key genes not only in neural crest cells, but also in the adult superior cervical ganglion and locus ceruleus. In addition to known genes we have identified novel differentially expressed genes and thus provide a valuable database for future studies. CONCLUSION: Loss of NET function during embryonic development in the mouse deregulates signaling pathways that are critically involved in neural crest formation and noradrenergic cell differentiation. The data further suggest deregulation of signaling pathways in the development and/or function of the NET-deficient peripheral, central and enteric nervous systems.


Subject(s)
Gene Expression Profiling , Neural Crest/metabolism , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine/metabolism , Animals , Biological Transport , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Gene Library , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/cytology , Neural Crest/embryology , Neurons/cytology , Neurons/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/physiology , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT3 , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
16.
Dev Neurosci ; 31(3): 202-11, 2009.
Article in English | MEDLINE | ID: mdl-19145072

ABSTRACT

Here we show that stem cell factor (SCF) signaling through its receptor, c-kit, is essential for the development of c-kit-expressing small- and medium-diameter primary sensory neurons. We used the W mouse, which is c-kit deficient and has a perinatal lethal phenotype due to a naturally occurring point mutation in the c-kit gene. In c-kit-null newborn mice, 52.5% of substance P immunoreactive and 31.4% of calcitonin gene-related peptide (CGRP) immunoreactive small- and medium-diameter sensory neurons were absent, whereas large-diameter sensory neurons were unaffected. Equivalent deficits occurred during embryogenesis. There was neither a developmental delay nor degeneration of differentiated neurons. We thus conclude that, in the absence of SCF signaling, neural crest-derived progenitors do not differentiate into c-kit-expressing visceral and somatic afferent neurons.


Subject(s)
Neurogenesis/physiology , Sensory Receptor Cells/cytology , Signal Transduction/physiology , Stem Cell Factor/metabolism , Animals , Base Sequence , Calcitonin Gene-Related Peptide/biosynthesis , Cell Differentiation , Fluorescent Antibody Technique , Ganglia, Spinal/embryology , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Mice, Mutant Strains , Mutation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-kit/genetics , Sensory Receptor Cells/metabolism , Substance P/biosynthesis
17.
J Vis Exp ; (15)2008 May 09.
Article in English | MEDLINE | ID: mdl-19066583

ABSTRACT

EPI-NCSC are remnants of the embryonic neural crest in an adult location, the bulge of hair follicles. They are multipotent stem cells that have the physiological property to generate a wide array of differentiated cell types, including neurons, nerve supporting cells, smooth muscle cells, bone/cartilage cells and melanocytes. EPI-NCSC are easily accessible in the hairy skin and can be isolated as a highly pure population of stem cells. This video provides a detailed protocol for preparing mouse EPI-NCSC cultures from whisker follicles. The whisker pad of an adult mouse is removed, and whisker follicles dissected. The follicles are then cut longitudinally and subsequently transversely above and below the bulge region. The bulge is removed from the collagen capsule and placed in a culture plate. EPI-NCSC start to emigrate from the bulge explants 3 to 4 days later.


Subject(s)
Cell Culture Techniques/methods , Multipotent Stem Cells/cytology , Neural Crest/cytology , Animals , Hair Follicle/cytology , Mice
18.
Stem Cell Rev ; 4(4): 256-60, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18712509

ABSTRACT

This article serves three purposes. We summarize current knowledge of the origin and characteristics of EPI-NCSC, review their application in a mouse model of spinal cord injury, and we present new data that highlight aspects of pluripotency of EPI-NCSC. EPI-NCSC are multipotent stem cells, which are derived from the embryonic neural crest and are located in the bulge of hair follicles. EPI-NCSC can undergo self-renewal and they are able to generate all major neural crest derivatives, including neurons, nerve supporting cells, smooth muscle cells, bone/cartilage cells and melanocytes. Despite their ectodermal origin, neural crest cells can also generate cell types that typically are derived from mesoderm. We were therefore interested in exploring aspects of EPI-NCSC pluripotency. We here show that EPI-NCSC can fuse with adult skeletal muscle fibers and that incorporated EPI-NCSC nuclei are functional. Furthermore, we show that adult skeletal muscle represents an environment conducive to long-term survival of neurogenic EPI-NCSC. Genes used to create induced pluripotent stem (iPS) cells are present in our EPI-NCSC longSAGE gene expression library. Here we have corroborated this notion by real-time PCR. Our results show similarities in the expression of Myc, Klf4, Sox2 and Lin28 genes between EPI-NCSC and embryonic stem cells (ESC). In contrast there were major differences in Nanog and Pou5f1 (Oct-4) expression levels between EPI-NCSC and ESC, possibly explaining why EPI-NCSC are not tumorigenic. Overall, as embryonic remnants in an adult location EPI-NCSC show several attractive characteristics for future cell replacement therapy and/or biomedical engineering: Due to their ability to migrate, EPI-NCSC can be isolated as a highly pure population of multipotent stem cells by minimally-invasive procedures. The cells can be expanded in vitro into millions of stem cells/progenitors and they share some characteristics with pluripotent stem cells without being tumorigenic. Since the patients' own EPI-NCSC could be used for autologous transplantation, this would avoid graft rejection.


Subject(s)
Cell Differentiation/physiology , Epidermal Cells , Multipotent Stem Cells/cytology , Neural Crest/cytology , Animals , Humans , Kruppel-Like Factor 4 , Mice , Multipotent Stem Cells/physiology , Multipotent Stem Cells/transplantation , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Neural Crest/physiology , Neurons/cytology , Neurons/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy
19.
Stem Cells ; 24(12): 2692-702, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16931771

ABSTRACT

Here, we report the first transcriptome for mouse epidermal neural crest stem cells (EPI-NCSC, formerly eNCSCs). In addition, our study resolves conflicting opinions in the literature by showing that EPI-NCSC are distinct from other types of skin-resident stem cells/progenitors. Finally, with the three gene profiles, we have established a foundation and provide a valuable resource for future mouse NCSC research. EPI-NCSC represent a novel type of multipotent adult stem cell that originates from the embryonic neural crest and resides in the bulge of hair follicles. We performed gene profiling by LongSAGE (long serial analysis of gene expression) with mRNA from EPI-NCSC, embryonic NCSC, and in vitro differentiated embryonic neural crest progeny. We have identified important differentially expressed genes, including novel genes and disease genes. Furthermore, using stringent criteria, we have defined an NCSC molecular signature that consists of a panel of 19 genes and is representative of both EPI-NCSC and NCSC. EPI-NCSC have characteristics that combine advantages of embryonic and adult stem cells. Similar to embryonic stem cells, EPI-NCSC have a high degree of innate plasticity, they can be isolated at high levels of purity, and they can be expanded in vitro. Similar to other types of adult stem cell, EPI-NCSC are readily accessible by minimal invasive procedure. Multipotent adult mammalian stem cells are of great interest because of their potential value in future cell replacement therapy by autologous transplantation, which avoids graft rejection.


Subject(s)
Epidermal Cells , Epidermis/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Antigens, CD34/immunology , Base Sequence , Biomarkers , Cells, Cultured , DNA-Binding Proteins/immunology , Dermis/cytology , Gene Expression Profiling , Gene Expression Regulation , Homeodomain Proteins/immunology , Mice , Mice, Inbred C57BL , Myosins/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Mol Cell Neurosci ; 32(1-2): 67-81, 2006.
Article in English | MEDLINE | ID: mdl-16626970

ABSTRACT

We have characterized in the contusion-lesioned murine spinal cord the behavior of acutely implanted epidermal neural crest stem cells (EPI-NCSC, formerly eNCSC). EPI-NCSC, a novel type of multipotent adult stem cell, are remnants of the embryonic neural crest. They reside in the bulge of hair follicles and have the ability to differentiate into all major neural crest derivatives (Sieber-Blum, M., Grim, M., Hu, Y.F., Szeder, V., 2004. Pluripotent neural crest stem cells in the adult hair follicle. Dev. Dyn. 231, 258-269). Grafted EPI-NCSC survived, integrated, and intermingled with host neurites in the lesioned spinal cord. EPI-NCSC were non-migratory. They did not proliferate and did not form tumors. Significant subsets expressed neuron-specific beta-III tubulin, the GABAergic marker glutamate decarboxylase 67 (GAD67), the oligodendrocyte marker, RIP, or myelin basic protein (MBP). Close physical association of non-neuronal EPI-NCSC with host neurites was observed. Glial fibrillary acidic protein (GFAP) immunofluorescence was not detected. Collectively, our data indicate that intraspinal EPI-NCSC demonstrate several desirable characteristics that may include local neural replacement and re-myelination.


Subject(s)
Multipotent Stem Cells/transplantation , Neural Crest/transplantation , Spinal Cord Injuries/therapy , Spinal Cord/cytology , Spinal Cord/physiology , Stem Cell Transplantation/methods , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Disease Models, Animal , Epidermal Cells , Epidermis/embryology , Epidermis/metabolism , Graft Survival/physiology , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Neurites/metabolism , Neurites/ultrastructure , Spinal Cord/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...