Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Fluids Barriers CNS ; 21(1): 43, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773599

ABSTRACT

The European Choroid plexus Scientific Forum (ECSF), held in Heidelberg, Germany between the 7th and 9th of November 2023, involved 21 speakers from eight countries. ECSF focused on discussing cutting-edge fundamental and medical research related to the development and functions of the choroid plexus and its implications for health, aging, and disease, including choroid plexus tumors. In addition to new findings in this expanding field, innovative approaches, animal models and 3D in vitro models were showcased to encourage further investigation into choroid plexus and cerebrospinal fluid roles.


Subject(s)
Choroid Plexus , Humans , Animals , Cerebrospinal Fluid , Europe , Choroid Plexus Neoplasms
2.
Science ; 372(6547): 1205-1209, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34112692

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse ventricular-subventricular zone (V-SVZ) undergo activation to generate neurons and some glia. Here we show that platelet-derived growth factor receptor beta (PDGFRß) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia. Selective deletion of PDGFRß in adult V-SVZ NSCs leads to their release from quiescence, uncovering gliogenic domains for different glial cell types. These domains are also recruited upon injury. We identify an intraventricular oligodendrocyte progenitor derived from NSCs inside the brain ventricles that contacts supraependymal axons. Together, our findings reveal that the adult V-SVZ contains spatial domains for gliogenesis, in addition to those for neurogenesis. These gliogenic NSC domains tend to be quiescent under homeostasis and may contribute to brain plasticity.


Subject(s)
Adult Stem Cells/physiology , Cerebral Ventricles/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neuroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Astrocytes/cytology , Astrocytes/physiology , Axons/physiology , Cell Differentiation , Cell Division , Cerebral Ventricles/cytology , Ependyma/cytology , Ependyma/physiology , Female , Gene Expression Profiling , Homeostasis , Lateral Ventricles/cytology , Male , Mice , Neurogenesis , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics
3.
Science ; 369(6500): 143-144, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32646987

Subject(s)
Brain , Organoids , Feces , Humans
4.
Cell Rep ; 26(2): 394-406.e5, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625322

ABSTRACT

The ventricular-subventricular zone (V-SVZ) harbors adult neural stem cells. V-SVZ neural stem cells exhibit features of astrocytes, have a regional identity, and depending on their location in the lateral or septal wall of the lateral ventricle, generate different types of neuronal and glial progeny. We performed large-scale single-cell RNA sequencing to provide a molecular atlas of cells from the lateral and septal adult V-SVZ of male and female mice. This revealed regional and sex differences among adult V-SVZ cells. We uncovered lineage potency bias at the single-cell level among lateral and septal wall astrocytes toward neurogenesis and oligodendrogenesis, respectively. Finally, we identified transcription factor co-expression modules marking key temporal steps in neurogenic and oligodendrocyte lineage progression. Our data suggest functionally important spatial diversity in neurogenesis and oligodendrogenesis in the adult brain and reveal molecular correlates of adult NSC dormancy and lineage specialization.


Subject(s)
Cell Lineage , Lateral Ventricles/cytology , Neural Stem Cells/cytology , Single-Cell Analysis/methods , Transcriptome , Animals , Astrocytes/cytology , Astrocytes/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , Organ Specificity
5.
Neuron ; 98(2): 246-248, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29673477

ABSTRACT

Obernier et al. (2018) show that the primary mode of division of adult ventricular-subventricular zone (V-SVZ) neural stem cells is symmetric, with the majority generating two non-stem cell progeny, and a minority self-renewing. This discovery has important implications for understanding stem cell dynamics and adult neurogenesis.


Subject(s)
Adult Stem Cells/physiology , Cell Division/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Adult , Animals , Cell Lineage/physiology , Humans
6.
Acta Neuropathol ; 135(3): 337-361, 2018 03.
Article in English | MEDLINE | ID: mdl-29368213

ABSTRACT

The barrier between the blood and the ventricular cerebrospinal fluid (CSF) is located at the choroid plexuses. At the interface between two circulating fluids, these richly vascularized veil-like structures display a peculiar morphology explained by their developmental origin, and fulfill several functions essential for CNS homeostasis. They form a neuroprotective barrier preventing the accumulation of noxious compounds into the CSF and brain, and secrete CSF, which participates in the maintenance of a stable CNS internal environment. The CSF circulation plays an important role in volume transmission within the developing and adult brain, and CSF compartments are key to the immune surveillance of the CNS. In these contexts, the choroid plexuses are an important source of biologically active molecules involved in brain development, stem cell proliferation and differentiation, and brain repair. By sensing both physiological changes in brain homeostasis and peripheral or central insults such as inflammation, they also act as sentinels for the CNS. Finally, their role in the control of immune cell traffic between the blood and the CSF confers on the choroid plexuses a function in neuroimmune regulation and implicates them in neuroinflammation. The choroid plexuses, therefore, deserve more attention while investigating the pathophysiology of CNS diseases and related comorbidities.


Subject(s)
Blood-Brain Barrier/anatomy & histology , Blood-Brain Barrier/metabolism , Cerebral Ventricles/anatomy & histology , Cerebral Ventricles/metabolism , Cerebrospinal Fluid/metabolism , Animals , Blood-Brain Barrier/pathology , Cerebral Ventricles/pathology , Humans , Neuroprotection/physiology
7.
Stem Cell Reports ; 8(5): 1421-1429, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28434940

ABSTRACT

Characterization of non-neoplastic and malignant human stem cell populations in their native state can provide new insights into gliomagenesis. Here we developed a purification strategy to directly isolate EGFR+/- populations from human germinal matrix (GM) and adult subventricular zone autopsy tissues, and from de novo glioblastoma (GBM) resections, enriching for cells capable of binding EGF ligand (LBEGFR+), and uniquely compared their functional and molecular properties. LBEGFR+ populations in both GM and GBM encompassed all sphere-forming cells and displayed proliferative stem cell properties in vitro. In xenografts, LBEGFR+ GBM cells showed robust tumor initiation and progression to high-grade, infiltrative gliomas. Whole-transcriptome sequencing analysis confirmed enrichment of proliferative pathways in both developing and neoplastic freshly isolated EGFR+ populations, and identified both unique and shared sets of genes. The ability to prospectively isolate stem cell populations using native ligand-binding capacity opens new doors onto understanding both normal human development and tumor cell biology.


Subject(s)
Brain Neoplasms/pathology , Cell Proliferation , Glioblastoma/pathology , Neoplastic Stem Cells/physiology , Neural Stem Cells/physiology , Animals , Brain Neoplasms/metabolism , Cell Separation/methods , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/metabolism , Humans , Male , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Neural Stem Cells/metabolism , Primary Cell Culture/methods , Transcriptome , Xenograft Model Antitumor Assays
8.
Cell Stem Cell ; 19(5): 643-652, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27452173

ABSTRACT

Specialized niches support the lifelong maintenance and function of tissue-specific stem cells. Adult neural stem cells in the ventricular-subventricular zone (V-SVZ) contact the cerebrospinal fluid (CSF), which flows through the lateral ventricles. A largely ignored component of the V-SVZ stem cell niche is the lateral ventricle choroid plexus (LVCP), a primary producer of CSF. Here we show that the LVCP, in addition to performing important homeostatic support functions, secretes factors that promote colony formation and proliferation of purified quiescent and activated V-SVZ stem cells and transit-amplifying cells. The functional effect of the LVCP secretome changes throughout the lifespan, with activated neural stem cells being especially sensitive to age-related changes. Transcriptome analysis identified multiple factors that recruit colony formation and highlights novel facets of LVCP function. Thus, the LVCP is a key niche compartment that translates physiological changes into molecular signals directly affecting neural stem cell behavior.


Subject(s)
Adult Stem Cells/cytology , Aging/physiology , Choroid Plexus/cytology , Neural Stem Cells/cytology , Signal Transduction , Stem Cell Niche , Adult Stem Cells/metabolism , Animals , Cell Proliferation , Choroid Plexus/metabolism , Clone Cells , Gene Expression Profiling , Lateral Ventricles/cytology , Mice , Neural Stem Cells/metabolism
9.
J Neurosci ; 35(11): 4528-39, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25788671

ABSTRACT

Adult neural stem cells reside in specialized niches. In the ventricular-subventricular zone (V-SVZ), quiescent neural stem cells (qNSCs) become activated (aNSCs), and generate transit amplifying cells (TACs), which give rise to neuroblasts that migrate to the olfactory bulb. The vasculature is an important component of the adult neural stem cell niche, but whether vascular cells in neurogenic areas are intrinsically different from those elsewhere in the brain is unknown. Moreover, the contribution of pericytes to the neural stem cell niche has not been defined. Here, we describe a rapid FACS purification strategy to simultaneously isolate primary endothelial cells and pericytes from brain microregions of nontransgenic mice using CD31 and CD13 as surface markers. We compared the effect of purified vascular cells from a neurogenic (V-SVZ) and non-neurogenic brain region (cortex) on the V-SVZ stem cell lineage in vitro. Endothelial and pericyte diffusible signals from both regions differentially promote the proliferation and neuronal differentiation of qNSCs, aNSCs, and TACs. Unexpectedly, diffusible cortical signals had the most potent effects on V-SVZ proliferation and neurogenesis, highlighting the intrinsic capacity of non-neurogenic vasculature to support stem cell behavior. Finally, we identify PlGF-2 as an endothelial-derived mitogen that promotes V-SVZ cell proliferation. This purification strategy provides a platform to define the functional and molecular contribution of vascular cells to stem cell niches and other brain regions under different physiological and pathological states.


Subject(s)
Adult Stem Cells/physiology , Cell Lineage/physiology , Cerebral Ventricles/physiology , Endothelium, Vascular/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Cell Proliferation/physiology , Cells, Cultured , Cerebral Ventricles/cytology , Endothelium, Vascular/cytology , Male , Mice
10.
Neuron ; 83(3): 507-9, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25102554

ABSTRACT

A major question in studying adult neurogenesis is the source and identity of molecules that regulate stem cells. In this issue of Neuron, uncover that endothelial-derived NT-3 acts as a mediator of quiescence in the V-SVZ adult neural stem cell niche.


Subject(s)
Endothelial Cells/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Neurotrophin 3/metabolism , Nitric Oxide/metabolism , Animals
11.
Neuron ; 82(3): 545-59, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811379

ABSTRACT

Adult neurogenic niches harbor quiescent neural stem cells; however, their in vivo identity has been elusive. Here, we prospectively isolate GFAP(+)CD133(+) (quiescent neural stem cells [qNSCs]) and GFAP(+)CD133(+)EGFR(+) (activated neural stem cells [aNSCs]) from the adult ventricular-subventricular zone. aNSCs are rapidly cycling, highly neurogenic in vivo, and enriched in colony-forming cells in vitro. In contrast, qNSCs are largely dormant in vivo, generate olfactory bulb interneurons with slower kinetics, and only rarely form colonies in vitro. Moreover, qNSCs are Nestin negative, a marker widely used for neural stem cells. Upon activation, qNSCs upregulate Nestin and EGFR and become highly proliferative. Notably, qNSCs and aNSCs can interconvert in vitro. Transcriptome analysis reveals that qNSCs share features with quiescent stem cells from other organs. Finally, small-molecule screening identified the GPCR ligands, S1P and PGD2, as factors that actively maintain the quiescent state of qNSCs.


Subject(s)
Adult Stem Cells/physiology , Lateral Ventricles/cytology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Animals , Astrocytes/physiology , Biomarkers/metabolism , Cell Separation/methods , Cells, Cultured , Humans , Mice , Mice, Transgenic , Prospective Studies , Transcriptome/genetics
12.
Curr Opin Neurobiol ; 23(6): 935-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24090877

ABSTRACT

Stem cells persist in specialized niches in the adult mammalian brain. Emerging findings highlight the complexity and heterogeneity of different compartments in the niche, as well as the presence of local signaling microdomains. Stem cell quiescence and activation are regulated not only by anchorage to the niche and diffusible signals, but also by biophysical properties, including fluid dynamics. Importantly, the adult neural stem cell niche integrates both local and systemic changes, reflecting the physiological state of the organism. Moreover niche signaling is bidirectional, with stem cells and their progeny and niche cells dynamically interacting with each other during homeostasis, regeneration and aging.


Subject(s)
Adult Stem Cells/cytology , Aging/physiology , Homeostasis/physiology , Nerve Regeneration/physiology , Neural Stem Cells/cytology , Stem Cell Niche/physiology , Adult Stem Cells/metabolism , Animals , Humans , Neural Stem Cells/metabolism , Neurogenesis/physiology
13.
Cell Stem Cell ; 8(5): 486-98, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21549325

ABSTRACT

Sphere-forming assays have been widely used to retrospectively identify stem cells based on their reported capacity to evaluate self-renewal and differentiation at the single-cell level in vitro. The discovery of markers that allow the prospective isolation of stem cells and their progeny from their in vivo niche allows the functional properties of purified populations to be defined. We provide a historical perspective of the evolution of the neurosphere assay and highlight limitations in the use of sphere-forming assays in the context of neurospheres. We discuss theoretical and technical considerations of experimental design and interpretation that surround the use of this assay with any tissue.


Subject(s)
Colony-Forming Units Assay , Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Survival , Humans , Research Design
14.
Cell Stem Cell ; 3(3): 279-88, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18786415

ABSTRACT

Stem cells reside in specialized niches that regulate their self-renewal and differentiation. The vasculature is emerging as an important component of stem cell niches. Here, we show that the adult subventricular zone (SVZ) neural stem cell niche contains an extensive planar vascular plexus that has specialized properties. Dividing stem cells and their transit-amplifying progeny are tightly apposed to SVZ blood vessels both during homeostasis and regeneration. They frequently contact the vasculature at sites that lack astrocyte endfeet and pericyte coverage, a modification of the blood-brain barrier unique to the SVZ. Moreover, regeneration often occurs at these sites. Finally, we find that circulating small molecules in the blood enter the SVZ. Thus, the vasculature is a key component of the adult SVZ neural stem cell niche, with SVZ stem cells and transit-amplifying cells uniquely poised to receive spatial cues and regulatory signals from diverse elements of the vascular system.


Subject(s)
Adult Stem Cells/cytology , Blood Vessels/cytology , Brain/cytology , Lateral Ventricles/cytology , Adult Stem Cells/physiology , Animals , Brain/blood supply , Brain/physiology , Cell Differentiation , Cell Division , Humans , Lateral Ventricles/blood supply , Lateral Ventricles/physiology , Mice , Mice, Inbred Strains
15.
Curr Opin Genet Dev ; 16(5): 518-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16919447

ABSTRACT

The mammalian epidermis is a highly accessible tissue in which to study the properties of adult stem cells. Global gene expression profiling has revealed new markers and regulators of the stem cell compartment. Although stem cells have the potential to differentiate into multiple lineages, their progeny follow a more restricted number of lineages in undamaged epidermis as a result of local microenvironmental cues. The response of the epidermis to a particular signal depends on signal strength and duration. Recent advances in the field have led to elucidation of the mechanisms by which stem cells are maintained and the pathways that interact with Wnt signalling to specify lineage choice as cells leave the stem cell compartment. This work has also yielded new insights into skin tumour development.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biomarkers , Cell Proliferation , Humans , Signal Transduction , Wnt Proteins/metabolism
16.
J Invest Dermatol ; 125(6): 1215-27, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354192

ABSTRACT

There are conflicting reports of the consequences of deleting beta1 integrins from the epidermis of transgenic mice. Epidermal thinning with normal differentiation and lack of inflammation has been observed; conversely, epidermal thickening, abnormal differentiation, and dermal fibrosis can occur. beta1 integrin deletion results in decreased epidermal proliferation, yet on wounding the proliferative defect is overcome. To distinguish primary from secondary consequences of beta1 integrin loss, we compared epidermal beta1 deletion at E14.5 via K5Cre and 4-hydroxy-tamoxifen induced deletion in adulthood via K14CreER. As reported previously, there was dermo-epidermal splitting, inflammation, reduced proliferation, and hair follicle and sebaceous gland loss in 30-d-old K5Cre beta1-null mice. These changes were not observed 30 d after beta1 integrin deletion in adult epidermis, however, and there were no changes in the hair follicle stem cell compartment. Deletion in adult epidermis revealed a previously unreported correlation between the level of beta1 integrins and proliferation in the interfollicular epidermis that was remarkably consistent with human epidermis. In addition, the number of melanocytes in interfollicular epidermis was greatly increased. Our results highlight the context-dependent effects of beta1 integrin deletion and suggest that inflammation may be responsible for some of the K5Cre beta1-null phenotype.


Subject(s)
Epidermis/physiology , Integrin beta1/genetics , Integrins/physiology , Skin Physiological Phenomena , Animals , Animals, Newborn , Cell Communication , Cell Differentiation , Cell Division , Epidermis/growth & development , Gene Deletion , Mice , Mice, Knockout , Skin Aging
17.
Dev Cell ; 9(1): 121-31, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15992546

ABSTRACT

Using K14deltaNbeta-cateninER transgenic mice, we show that short-term, low-level beta-catenin activation stimulates de novo hair follicle formation from sebaceous glands and interfollicular epidermis, while only sustained, high-level activation induces new follicles from preexisting follicles. The Hedgehog pathway is upregulated by beta-catenin activation, and inhibition of Hedgehog signaling converts the low beta-catenin phenotype to wild-type epidermis and the high phenotype to low. beta-catenin-induced follicles contain clonogenic keratinocytes that express bulge markers; the follicles induce dermal papillae and provide a niche for melanocytes, and they undergo 4OHT-dependent cycles of growth and regression. New follicles induced in interfollicular epidermis are derived from that cellular compartment and not through bulge stem cell migration or division. These results demonstrate the remarkable capacity of adult epidermis to be reprogrammed by titrating beta-catenin and Hedgehog signal strength and establish that cells from interfollicular epidermis can acquire certain characteristics of bulge stem cells.


Subject(s)
Cytoskeletal Proteins/metabolism , Epidermal Cells , Hair Follicle/cytology , Stem Cells/cytology , Trans-Activators/metabolism , Animals , Cell Differentiation , Cytoskeletal Proteins/genetics , Epidermis/drug effects , Epidermis/metabolism , Female , Gene Dosage , Hair Follicle/drug effects , Hair Follicle/metabolism , Hedgehog Proteins , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Sebaceous Glands/cytology , Sebaceous Glands/drug effects , Sebaceous Glands/metabolism , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Trans-Activators/genetics , Transgenes , beta Catenin
18.
J Invest Dermatol ; 123(3): 503-15, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15304090

ABSTRACT

Epidermal activation of Erk MAPK is observed in human psoriatic lesions and in a mouse model of psoriasis in which beta1 integrins are expressed in the suprabasal epidermal layers. Constitutive activation of the upstream kinase MEK1 causes hyperproliferation and perturbed differentiation of human keratinocytes in culture. It is not known, however, whether Erk activation in differentiating keratinocytes is sufficient to trigger hyperproliferation of basal keratinocytes and a skin inflammatory infiltrate. To investigate this, we expressed constitutively active MEK1 in the suprabasal epidermal layers of transgenic mice. Proliferation in the epidermal basal layer was stimulated and epidermal terminal differentiation was perturbed. Some older mice also developed papillomas. There was a large increase in T lymphocytes, dendritic cells, and neutrophils in the skin. The effects of suprabasal MEK1 on basal keratinocytes and leukocytes, cells that were transgene negative, suggested that MEK1 activity might stimulate cytokine release. Transgenic keratinocytes expressed elevated IL-1alpha and crossing the mice with mice overexpressing the IL-1 receptor in the epidermal basal layer led to exacerbated hyperproliferation and inflammation. These data suggest that activation of MEK1 downstream of beta1 integrins plays an important role in epidermal hyperproliferation and skin inflammation.


Subject(s)
Epidermis/physiopathology , Keratinocytes/pathology , Keratosis/physiopathology , Mitogen-Activated Protein Kinase Kinases/genetics , Animals , Biomarkers , Cell Communication/immunology , Cell Differentiation/physiology , Cell Division/physiology , Epidermis/immunology , Epidermis/pathology , Extremities , Female , Interleukin-1/genetics , Keratinocytes/immunology , Keratosis/immunology , Keratosis/pathology , MAP Kinase Kinase 1 , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Papilloma/immunology , Papilloma/pathology , Papilloma/physiopathology , Phenotype , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology
19.
Development ; 130(21): 5241-55, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12954714

ABSTRACT

Mammalian epidermis is maintained by stem cells that have the ability to self-renew and generate daughter cells that differentiate along the lineages of the hair follicles, interfollicular epidermis and sebaceous gland. As stem cells divide infrequently in adult mouse epidermis, they can be visualised as DNA label-retaining cells (LRC). With whole-mount labelling, we can examine large areas of interfollicular epidermis and many hair follicles simultaneously, enabling us to evaluate stem cell markers and examine the effects of different stimuli on the LRC population. LRC are not confined to the hair follicle, but also lie in sebaceous glands and interfollicular epidermis. LRC reside throughout the permanent region of the hair follicle, where they express keratin 15 and lie in a region of high alpha6beta4 integrin expression. LRC are not significantly depleted by successive hair growth cycles. They can, nevertheless, be stimulated to divide by treatment with phorbol ester, resulting in near complete loss of LRC within 12 days. Activation of Myc stimulates epidermal proliferation without depleting LRC and induces differentiation of sebocytes within the interfollicular epidermis. Expression of N-terminally truncated Lef1 to block beta-catenin signalling induces transdifferentiation of hair follicles into interfollicular epidermis and sebocytes and causes loss of LRC primarily through proliferation. We conclude that LRC are more sensitive to some proliferative stimuli than others and that changes in lineage can occur with or without recruitment of LRC into cycle.


Subject(s)
Cell Division/physiology , Cell Lineage , Epidermal Cells , Staining and Labeling/methods , Stem Cells/physiology , Animals , Biomarkers , Bromodeoxyuridine/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epidermis/metabolism , Hair/cytology , Hair/physiology , Histocytological Preparation Techniques , Lymphoid Enhancer-Binding Factor 1 , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...