Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Gene Expr Patterns ; 23-24: 32-44, 2017 01.
Article in English | MEDLINE | ID: mdl-28288836

ABSTRACT

Rod and cone photoreceptors are photosensitive cells in the retina that convert light to electrical signals that are transmitted to visual processing centres in the brain. During development, cones and rods are generated from a common pool of multipotent retinal progenitor cells (RPCs) that also give rise to other retinal cell types. Cones and rods differentiate in two distinct waves, peaking in mid-embryogenesis and the early postnatal period, respectively. As RPCs transition from making cones to generating rods, there are changes in the expression profiles of genes involved in photoreceptor cell fate specification and differentiation. To better understand the temporal transition from cone to rod genesis, we assessed the timing of onset and offset of expression of a panel of 11 transcription factors and 7 non-transcription factors known to function in photoreceptor development, examining their expression between embryonic day (E) 12.5 and postnatal day (P) 60. Transcription factor expression in the photoreceptor layer was observed as early as E12.5, beginning with Crx, Otx2, Rorb, Neurod1 and Prdm1 expression, followed at E15.5 with the expression of Thrb, Neurog1, Sall3 and Rxrg expression, and at P0 by Nrl and Nr2e3 expression. Of the non-transcription factors, peanut agglutinin lectin staining and cone arrestin protein were observed as early as E15.5 in the developing outer nuclear layer, while transcripts for the cone opsins Opn1mw and Opn1sw and Recoverin protein were detected in photoreceptors by P0. In contrast, Opn1mw and Opn1sw protein were not observed in cones until P7, when rod-specific Gnat1 transcripts and rhodopsin protein were also detected. We have thus identified four transitory stages during murine retina photoreceptor differentiation marked by the period of onset of expression of new photoreceptor lineage genes. By characterizing these stages, we have clarified the dynamic nature of gene expression during the period when photoreceptor identities are progressively acquired during development.


Subject(s)
Photoreceptor Cells, Vertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Male , Mice , Retina/cytology , Retina/embryology , Retina/metabolism , Transcriptome
2.
Proc Natl Acad Sci U S A ; 114(9): E1707-E1716, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28137843

ABSTRACT

AlphaB-crystallin (αBC) is a small heat shock protein that is constitutively expressed by peripheral nervous system (PNS) axons and Schwann cells. To determine what role this crystallin plays after peripheral nerve damage, we found that loss of αBC impaired remyelination, which correlated with a reduced presence of myelinating Schwann cells and increased numbers of nonmyelinating Schwann cells. The heat shock protein also seems to regulate the cross-talk between Schwann cells and axons, because expected changes in neuregulin levels and ErbB2 receptor expression after PNS injury were disrupted in the absence of αBC. Such dysregulations led to defects in conduction velocity and motor and sensory functions that could be rescued with therapeutic application of the heat shock protein in vivo. Altogether, these findings show that αBC plays an important role in regulating Wallerian degeneration and remyelination after PNS injury.


Subject(s)
Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Remyelination/physiology , alpha-Crystallin B Chain/metabolism , Animals , Axons/metabolism , Axons/physiology , Female , Heat-Shock Proteins/metabolism , Mice , Myelin Sheath/metabolism , Myelin Sheath/physiology , Peripheral Nervous System/metabolism , Peripheral Nervous System/physiopathology , Receptor, ErbB-2/metabolism , Schwann Cells/physiology
3.
Exp Neurol ; 254: 168-79, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440805

ABSTRACT

Previous work has shown that infusion of skin-derived precursors pre-differentiated into Schwann cells (SKP-SCs) can remyelinate injured and regenerating axons, and improve indices of axonal regeneration and electrophysiological parameters in rodents. We hypothesized that SKP-SC therapy would improve behavioral outcomes following nerve injury repair and tested this in a pre-clinical trial in 90 rats. A model of sciatic nerve injury and acellular graft repair was used to compare injected SKP-SCs to nerve-derived Schwann cells or media, and each was compared to the gold standard nerve isograft repair. In a second experiment, rats underwent right tibial nerve transection and received either acute or delayed direct nerve repair, with injections of either 1) SKP-SCs distal to the repair site, 2) carrier medium alone, or 3) dead SKP-SCs, and were followed for 4, 8 or 17weeks. For delayed repairs, both transected nerve ends were capped and repaired 11weeks later, along with injections of cells or media as above, and followed for 9 additional weeks (total of 20weeks). Rats were serially tested for skilled locomotion and a slip ratio was calculated for the horizontal ladder-rung and tapered beam tasks. Immediately after nerve injury and with chronic denervation, slip ratios were dramatically elevated. In the GRAFT repair study, the SKP-SC treated rats showed statistically significant improvement in ladder rung as compared to all other groups, and exhibited the greatest similarity to the sham controls on the tapered beam by study termination. In the ACUTE repair arm, the SKP-SC group showed marked improvement in ladder rung slip ratio as early as 5weeks after surgery, which was sustained for the duration of the experiment. Groups that received media and dead SKP-SCs improved with significantly slower progression. In the DELAYED repair arm, the SKP-SC group became significantly better than other groups 7weeks after the repair, while the media and the dead SKP-SCs showed no significant improvement in slip ratios. On histomorphometrical analysis, SKP-SC group showed significantly increased mean axon counts while the percent myelin debris was significantly lower at both 4 and 8weeks, suggesting that a less inhibitory micro-environment may have contributed to accelerated axonal regeneration. For delayed repair, mean axon counts were significantly higher in the SKP-SC group. Compound action potential amplitudes and muscle weights were also improved by cell therapy. In conclusion, SKP-SC therapy improves behavioral recovery after acute, chronic and nerve graft repair beyond the current standard of microsurgical nerve repair.


Subject(s)
Adult Stem Cells/transplantation , Dermis/cytology , Nerve Regeneration/physiology , Peripheral Nerve Injuries/therapy , Schwann Cells/transplantation , Stem Cell Transplantation/methods , Acute Disease , Adult Stem Cells/cytology , Animals , Animals, Newborn , Dermis/innervation , Female , Male , Motor Activity , Muscle, Skeletal/innervation , Peripheral Nerve Injuries/pathology , Primary Cell Culture , Rats , Rats, Inbred Lew , Schwann Cells/cytology , Sciatic Nerve/cytology , Time Factors
4.
Mol Pharm ; 9(5): 1521-8, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22480259

ABSTRACT

This study reports the use of a nonviral expression system based on polyamidoamine dendrimers for time-restricted neurotrophin overproduction in mesenchymal stem cells and skin precursor-derived Schwann cells. The dendrimers were used to deliver plasmids for brain-derived neurotrophic factor (BDNF) or neurotrophin-3 (NT-3) expression in both rodent and human stem cells, and the timelines of expression were studied. We have found that, despite the fact that transfection efficiencies and protein expression levels were comparable, dendrimer-driven expression in human mesenchymal stem cells was characterized by a more rapid decline compared to rodent cells. Transient expression systems can be beneficial for some neurotrophins, which were earlier reported to cause unwanted side effects in virus-based long-term expression models. Nonviral neurotrophin expression is a biologically safe and accessible alternative to increase the therapeutic potential of autologous adult stem cells and stem cell-derived functional differentiated cells.


Subject(s)
Dendrimers/chemistry , Nerve Growth Factors/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chondrogenesis/genetics , Chondrogenesis/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nerve Growth Factors/genetics , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Plasmids/genetics , Transfection/methods
5.
Stem Cells ; 28(11): 2027-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20848654

ABSTRACT

Skin-derived precursors (SKPs) are multipotent dermal stem cells that reside within a hair follicle niche and that share properties with embryonic neural crest precursors. Here, we have asked whether SKPs and their endogenous dermal precursors originate from the neural crest or whether, like the dermis itself, they originate from multiple developmental origins. To do this, we used two different mouse Cre lines that allow us to perform lineage tracing: Wnt1-cre, which targets cells deriving from the neural crest, and Myf5-cre, which targets cells of a somite origin. By crossing these Cre lines to reporter mice, we show that the endogenous follicle-associated dermal precursors in the face derive from the neural crest, and those in the dorsal trunk derive from the somites, as do the SKPs they generate. Despite these different developmental origins, SKPs from these two locations are functionally similar, even with regard to their ability to differentiate into Schwann cells, a cell type only thought to be generated from the neural crest. Analysis of global gene expression using microarrays confirmed that facial and dorsal SKPs exhibit a very high degree of similarity, and that they are also very similar to SKPs derived from ventral dermis, which has a lateral plate origin. However, these developmentally distinct SKPs also retain differential expression of a small number of genes that reflect their developmental origins. Thus, an adult neural crest-like dermal precursor can be generated from a non-neural crest origin, a finding with broad implications for the many neuroendocrine cells in the body.


Subject(s)
Neural Crest/cytology , Skin/cytology , Stem Cells/cytology , Animals , Computational Biology , Hair Follicle/cytology , Immunohistochemistry , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Tissue Culture Techniques
6.
Cell Stem Cell ; 5(6): 610-23, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19951689

ABSTRACT

Despite the remarkable regenerative capacity of mammalian skin, an adult dermal stem cell has not yet been identified. Here, we investigated whether skin-derived precursors (SKPs) might fulfill such a role. We show that SKPs derive from Sox2(+) hair follicle dermal cells and that these two cell populations are similar with regard to their transcriptome and functional properties. Both clonal SKPs and endogenous Sox2(+) cells induce hair morphogenesis, differentiate into dermal cell types, and home to a hair follicle niche upon transplantation. Moreover, hair follicle-derived SKPs self-renew, maintain their multipotency, and serially reconstitute hair follicles. Finally, grafting experiments show that follicle-associated dermal cells move out of their niche to contribute cells for dermal maintenance and wound-healing. Thus, SKPs derive from Sox2(+) follicle-associated dermal precursors and display functional properties predicted of a dermal stem cell, contributing to dermal maintenance, wound-healing, and hair follicle morphogenesis.


Subject(s)
Adult Stem Cells/metabolism , Hair Follicle/cytology , Neurons/cytology , SOXB1 Transcription Factors/biosynthesis , Stem Cell Niche/cytology , Adult Stem Cells/cytology , Animals , Animals, Newborn , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mice , Mice, SCID , Mice, Transgenic , Morphogenesis , Rats , Rats, Sprague-Dawley , Regeneration , Stem Cell Transplantation , Wound Healing
7.
Toxicol In Vitro ; 23(8): 1522-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19720137

ABSTRACT

In the present study, we investigated whether precursor cells isolated from the dermis of infant human foreskin are capable to differentiate into hepatocyte-like cells upon sequential and gradual exposure to hepatogenic factors [fibroblast growth factor (FGF)-4, hepatocyte growth factor (HGF), insulin-transferrin-selenite (ITS), dexamethasone and oncostatin M (OSM)], mimicking the liver embryogenesis in vivo. Undifferentiated human skin-derived precursors (hSKP) are characterized by a fibroblast-like shape. Yet, they already express typical hepatic proteins, including cytokeratin (CK)-18, hepatocyte nuclear factor (HNF)-4 and HNF-1alpha. Microarray analysis further reveals gene expression of (i) the stemness markers nestin, POU5F1 (OCT-4), telomerase reverse transcriptase (TERT) and thymocyte differentiation antigen (THY)-1, (ii) biliary CK14 and CK19, (iii) biliary/foetal hepatic connexin (Cx)-43, and (iv) adult hepatic CK18, HNF-4 and HNF-1alpha. Upon differentiation, cells undergo morphological and phenotypic changes. As such, hSKP adopt a more polygonal-to-cuboidal cell shape. At the protein level, Cx43 expression is downregulated whereas typical hepatic markers, including alfa-foetoprotein (AFP), prealbumin (TTR) and albumin (ALB), become expressed in accordance to in vivo patterns observed during hepatogenesis. In conclusion, these data show for the first time that hSKP are capable to "trans" differentiate into hepatocyte-like cells upon mimicking the in vivo micro-environment of developing hepatocytes in vitro.


Subject(s)
Cell Differentiation/drug effects , Cytokines/pharmacology , Growth Substances/pharmacology , Hepatocytes/cytology , Liver/embryology , Skin/cytology , Stem Cells/cytology , Adult , Humans , Phenotype
8.
Cell Stem Cell ; 5(1): 64-75, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19570515

ABSTRACT

The cellular mechanisms that regulate the maintenance of adult tissue stem cells are still largely unknown. We show here that the p53 family member, TAp63, is essential for maintenance of epidermal and dermal precursors and that, in its absence, these precursors senesce and skin ages prematurely. Specifically, we have developed a TAp63 conditional knockout mouse and used it to ablate TAp63 in the germline (TAp63(-/-)) or in K14-expressing cells in the basal layer of the epidermis (TAp63(fl/fl);K14cre+). TAp63(-/-) mice age prematurely and develop blisters, skin ulcerations, senescence of hair follicle-associated dermal and epidermal cells, and decreased hair morphogenesis. These phenotypes are likely due to loss of TAp63 in dermal and epidermal precursors since both cell types show defective proliferation, early senescence, and genomic instability. These data indicate that TAp63 serves to maintain adult skin stem cells by regulating cellular senescence and genomic stability, thereby preventing premature tissue aging.


Subject(s)
Adult Stem Cells/physiology , Aging, Premature/etiology , Dermis/cytology , Epidermal Cells , Phosphoproteins/genetics , Phosphoproteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Adult Stem Cells/cytology , Aging, Premature/pathology , Animals , Cellular Senescence , DNA Damage , Genes, p53 , Genomic Instability , Hair Follicle/cytology , Keratinocytes/cytology , Mice , Mice, Knockout , Skin Aging/genetics , Skin Aging/pathology , Wound Healing/genetics
9.
Stem Cells Dev ; 18(6): 893-906, 2009.
Article in English | MEDLINE | ID: mdl-18834279

ABSTRACT

Skin-derived precursors (SKPs) are multipotent dermal precursors that share similarities with neural crest stem cells and that can give rise to peripheral neural and some mesodermal cell types, such as adipocytes. Here, we have asked whether rodent or human SKPs can generate other mesenchymally derived cell types, with a particular focus on osteocytes and chondrocytes. In culture, rodent and human foreskin-derived SKPs differentiated into alkaline-positive, collagen type-1-positive, mineralizing osteocytes, and into collagen type-II-positive chondrocytes that secreted chondrocyte-specific proteoglycans. Clonal analysis demonstrated that SKPs efficiently generated these skeletogenic cell types, and that they were multipotent with regard to the osteogenic and chondrogenic lineages. To ask if SKPs could generate these same lineages in vivo, genetically tagged, undifferentiated rat SKPs were transplanted into a tibial bone fracture model. Over the ensuing 6 weeks, many of the transplanted cells survived within the bone callus, where they were morphologically and phenotypically similar to the endogenous mesenchymal/osteogenic cells. Moreover, some transplanted cells adopted a mature osteocyte phenotype and integrated into the newly formed bone. Some transplanted cells also differentiated into chondrocytes and into smooth muscle cells and/or pericytes that were associated with blood vessels. Thus, both rodent and human SKPs generate skeletogenic cell types in culture, and the injured bone environment is sufficient to instruct SKPs to differentiate down an osteogenic lineage, in a fashion similar to the endogenous mesenchymal precursors.


Subject(s)
Bone and Bones/pathology , Cell Differentiation , Skin/cytology , Stem Cells/cytology , Wound Healing , Animals , Cell Lineage , Cells, Cultured , Chickens , Chondrocytes/cytology , Chondrogenesis , Humans , Infant, Newborn , Mesoderm/cytology , Mice , Multipotent Stem Cells/cytology , Osteocytes/cytology , Osteogenesis , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation
10.
Neuron ; 59(5): 708-21, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18786355

ABSTRACT

The genetic mechanisms that regulate neurodegeneration are only poorly understood. We show that the loss of one allele of the p53 family member, p73, makes mice susceptible to neurodegeneration as a consequence of aging or Alzheimer's disease (AD). Behavioral analyses demonstrated that old, but not young, p73+/- mice displayed reduced motor and cognitive function, CNS atrophy, and neuronal degeneration. Unexpectedly, brains of aged p73+/- mice demonstrated dramatic accumulations of phospho-tau (P-tau)-positive filaments. Moreover, when crossed to a mouse model of AD expressing a mutant amyloid precursor protein, brains of these mice showed neuronal degeneration and early and robust formation of tangle-like structures containing P-tau. The increase in P-tau was likely mediated by JNK; in p73+/- neurons, the activity of the p73 target JNK was enhanced, and JNK regulated P-tau levels. Thus, p73 is essential for preventing neurodegeneration, and haploinsufficiency for p73 may be a susceptibility factor for AD and other neurodegenerative disorders.


Subject(s)
Aging , Alzheimer Disease/metabolism , DNA-Binding Proteins/physiology , Neurodegenerative Diseases/etiology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , tau Proteins/metabolism , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Behavior, Animal , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Models, Animal , Embryo, Mammalian , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Flow Cytometry , Galactosides/metabolism , MAP Kinase Kinase 4/metabolism , Magnetic Resonance Imaging/methods , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Microglia/pathology , Mutation/genetics , Neurodegenerative Diseases/pathology , Nuclear Proteins/genetics , Phosphopyruvate Hydratase/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
11.
J Mol Cell Cardiol ; 45(5): 694-702, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18718475

ABSTRACT

Nestin-expressing cells were identified in the normal rat heart characterized by a small cell body and numerous processes and following an ischemic insult migrated to the infarct region. The present study was undertaken to identify the phenotype, origin and biological role of nestin-expressing cells during reparative fibrosis. A neural stem cell phenotype was identified based on musashi-1 expression, growth as a neurosphere, and differentiation to a neuronal cell. Using the Wnt1-cre; Z/EG transgenic mouse model, which expresses EGFP in embryologically-derived neural crest cells, the reporter signal was detected in nestin-expressing cells residing in the heart. In infarcted human hearts, nestin-expressing cells were detected in the viable myocardium and the scar and morphologically analogous to the population identified in the rat heart. Following either an ischemic insult or the acute administration of 6-hydroxydopamine, sympathetic sprouting was dependent on the physical association of neurofilament-M immunoreactive fibres with nestin-positive processes emanating from neural stem cells. To specifically study the biological role of the subpopulation in the infarct region, neural stem cells were isolated from the scar, fluorescently labelled and transplanted in the heart of 3-day post-MI rats. Injected scar-derived neural stem cells migrated to the infarct region and were used as a substrate for de novo blood vessel formation. These data have demonstrated that the heart contains a resident population of neural stem cells derived from the neural crest and participate in reparative fibrosis. Their manipulation could provide an alternative approach to ameliorate the healing process following ischemic injury.


Subject(s)
Heart/physiology , Neovascularization, Physiologic , Animals , Humans , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Neural Crest/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Oxidopamine/pharmacology , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism
12.
Nat Protoc ; 1(6): 2803-12, 2006.
Article in English | MEDLINE | ID: mdl-17406538

ABSTRACT

This protocol describes methods of isolating skin-derived precursors (SKPs) from rodent and human skin, and for generating and enriching Schwann cells from rodent SKPs. SKPs are isolated as a population of non-adherent cells from the dermis that proliferate and self-renew as floating spheres in response to fibroblast growth factor 2 (FGF2) and epidermal growth factor (EGF). Their differentiation into Schwann cells and subsequent enrichment of these differentiated progeny involves culturing SKPs as adherent cells in the absence of FGF2 and EGF, but in the presence of neuregulins, and then mechanically isolating the Schwann cell colonies using cloning cylinders. Methods for expanding and characterizing these Schwann cells are provided. Generation of primary SKPs takes approximately 2 weeks, while differentiation of Schwann cells requires an additional 4-6 weeks.


Subject(s)
Cell Culture Techniques , Multipotent Stem Cells/cytology , Schwann Cells/cytology , Skin/cytology , Animals , Cell Differentiation , Humans , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...