Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 134(12): 2873-2882, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24999588

ABSTRACT

Chemotherapy has severe side effects in normal rapidly proliferating organs, such as hair follicles, and causes massive apoptosis in hair matrix keratinocytes followed by hair loss. To define the molecular signature of hair follicle response to chemotherapy, human scalp hair follicles cultured ex vivo were treated with doxorubicin (DXR), and global microarray analysis was performed 3 hours after treatment. Microarray data revealed changes in expression of 504 genes in DXR-treated hair follicles versus controls. Among these genes, upregulations of several tumor necrosis factor family of apoptotic receptors (FAS, TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) receptors 1/2), as well as of a large number of keratin-associated protein genes, were seen after DXR treatment. Hair follicle apoptosis induced by DXR was significantly inhibited by either TRAIL-neutralizing antibody or caspase-8 inhibitor, thus suggesting a previously unreported role for TRAIL receptor signaling in mediating DXR-induced hair loss. These data demonstrate that the early phase of the hair follicle response to DXR includes upregulation of apoptosis-associated markers, as well as substantial reorganization of the terminal differentiation programs in hair follicle keratinocytes. These data provide an important platform for further studies toward the design of effective approaches for the management of chemotherapy-induced hair loss.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Doxorubicin/pharmacology , Hair Follicle/cytology , Alopecia/chemically induced , Alopecia/metabolism , Alopecia/pathology , Antineoplastic Agents/adverse effects , Caspase 8/drug effects , Caspase 8/metabolism , Cells, Cultured , DNA Damage/drug effects , Doxorubicin/adverse effects , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , fas Receptor/drug effects , fas Receptor/metabolism
2.
J Invest Dermatol ; 134(3): 827-837, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24126843

ABSTRACT

Bone morphogenetic protein (BMP) signaling plays a key role in the control of skin development and postnatal remodeling by regulating keratinocyte proliferation, differentiation, and apoptosis. To study the role of BMPs in wound-induced epidermal repair, we used transgenic mice overexpressing the BMP downstream component Smad1 under the control of a K14 promoter as an in vivo model, as well as ex vivo and in vitro assays. K14-caSmad1 (transgenic mice overexpressing a constitutively active form of Smad1 under K14 promoter) mice exhibited retarded wound healing associated with significant inhibition of proliferation and increased apoptosis in healing wound epithelium. Furthermore, microarray and quantitative real-time reverse-transcriptase-PCR (qRT-PCR) analyses revealed decreased expression of a number of cytoskeletal/cell motility-associated genes including wound-associated keratins (Krt16, Krt17) and Myosin VA (Myo5a), in the epidermis of K14-caSmad1 mice versus wild-type (WT) controls during wound healing. BMP treatment significantly inhibited keratinocyte migration ex vivo, and primary keratinocytes of K14-caSmad1 mice showed retarded migration compared with WT controls. Finally, small interfering RNA (siRNA)-mediated silencing of BMPR-1B in primary mouse keratinocytes accelerated cell migration and was associated with increased expression of Krt16, Krt17, and Myo5a compared with controls. Thus, this study demonstrates that BMPs inhibit keratinocyte proliferation, cytoskeletal organization, and migration in regenerating skin epithelium during wound healing, and raises a possibility for using BMP antagonists for the management of chronic wounds.


Subject(s)
Apoptosis/physiology , Bone Morphogenetic Proteins/metabolism , Epidermis/physiology , Keratinocytes/physiology , Signal Transduction/physiology , Wound Healing/physiology , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Epidermal Cells , Humans , Keratin-14/genetics , Keratinocytes/cytology , Mice , Mice, Inbred Strains , Mice, Transgenic , RNA, Small Interfering/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism
3.
Development ; 141(1): 101-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24346698

ABSTRACT

Chromatin structural states and their remodelling, including higher-order chromatin folding and three-dimensional (3D) genome organisation, play an important role in the control of gene expression. The role of 3D genome organisation in the control and execution of lineage-specific transcription programmes during the development and differentiation of multipotent stem cells into specialised cell types remains poorly understood. Here, we show that substantial remodelling of the higher-order chromatin structure of the epidermal differentiation complex (EDC), a keratinocyte lineage-specific gene locus on mouse chromosome 3, occurs during epidermal morphogenesis. During epidermal development, the locus relocates away from the nuclear periphery towards the nuclear interior into a compartment enriched in SC35-positive nuclear speckles. Relocation of the EDC locus occurs prior to the full activation of EDC genes involved in controlling terminal keratinocyte differentiation and is a lineage-specific, developmentally regulated event controlled by transcription factor p63, a master regulator of epidermal development. We also show that, in epidermal progenitor cells, p63 directly regulates the expression of the ATP-dependent chromatin remodeller Brg1, which binds to distinct domains within the EDC and is required for relocation of the EDC towards the nuclear interior. Furthermore, Brg1 also regulates gene expression within the EDC locus during epidermal morphogenesis. Thus, p63 and its direct target Brg1 play an essential role in remodelling the higher-order chromatin structure of the EDC and in the specific positioning of this locus within the landscape of the 3D nuclear space, as required for the efficient expression of EDC genes in epidermal progenitor cells during skin development.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , Multipotent Stem Cells/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/metabolism , DNA Helicases/genetics , Epidermal Cells , Epidermis/embryology , Epidermis/metabolism , GA-Binding Protein Transcription Factor/genetics , Gene Expression Regulation, Developmental , Keratinocytes/cytology , Keratinocytes/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Binding , Protein Folding , RNA Interference , RNA, Small Interfering , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic
4.
Development ; 138(22): 4843-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22028024

ABSTRACT

The Lhx2 transcription factor plays essential roles in morphogenesis and patterning of ectodermal derivatives as well as in controlling stem cell activity. Here, we show that during murine skin morphogenesis, Lhx2 is expressed in the hair follicle (HF) buds, whereas in postnatal telogen HFs Lhx2(+) cells reside in the stem cell-enriched epithelial compartments (bulge, secondary hair germ) and co-express selected stem cell markers (Sox9, Tcf4 and Lgr5). Remarkably, Lhx2(+) cells represent the vast majority of cells in the bulge and secondary hair germ that proliferate in response to skin injury. This is functionally important, as wound re-epithelization is significantly retarded in heterozygous Lhx2 knockout (+/-) mice, whereas anagen onset in the HFs located closely to the wound is accelerated compared with wild-type mice. Cell proliferation in the bulge and the number of Sox9(+) and Tcf4(+) cells in the HFs closely adjacent to the wound in Lhx2(+/-) mice are decreased in comparison with wild-type controls, whereas expression of Lgr5 and cell proliferation in the secondary hair germ are increased. Furthermore, acceleration of wound-induced anagen development in Lhx2(+/-) mice is inhibited by administration of Lgr5 siRNA. Finally, Chip-on-chip/ChIP-qPCR and reporter assay analyses identified Sox9, Tcf4 and Lgr5 as direct Lhx2 targets in keratinocytes. These data strongly suggest that Lhx2 positively regulates Sox9 and Tcf4 in the bulge cells, and promotes wound re-epithelization, whereas it simultaneously negatively regulates Lgr5 in the secondary hair germ and inhibits HF cycling. Thus, Lhx2 operates as an important regulator of epithelial stem cell activity in the skin response to injury.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Epidermis/physiology , Hair Follicle/metabolism , LIM-Homeodomain Proteins/physiology , Receptors, G-Protein-Coupled/genetics , Regeneration/genetics , SOX9 Transcription Factor/genetics , Stem Cells/physiology , Transcription Factors/physiology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Embryo, Mammalian , Epidermis/injuries , Epidermis/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Hair Follicle/cytology , Humans , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , RNA, Small Interfering/pharmacology , Receptors, G-Protein-Coupled/metabolism , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Transcription Factor 4 , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing/drug effects , Wound Healing/genetics
5.
J Cell Biol ; 194(6): 825-39, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21930775

ABSTRACT

During development, multipotent progenitor cells establish tissue-specific programs of gene expression. In this paper, we show that p63 transcription factor, a master regulator of epidermal morphogenesis, executes its function in part by directly regulating expression of the genome organizer Satb1 in progenitor cells. p63 binds to a proximal regulatory region of the Satb1 gene, and p63 ablation results in marked reduction in the Satb1 expression levels in the epidermis. Satb1(-/-) mice show impaired epidermal morphology. In Satb1-null epidermis, chromatin architecture of the epidermal differentiation complex locus containing genes associated with epidermal differentiation is altered primarily at its central domain, where Satb1 binding was confirmed by chromatin immunoprecipitation-on-chip analysis. Furthermore, genes within this domain fail to be properly activated upon terminal differentiation. Satb1 expression in p63(+/-) skin explants treated with p63 small interfering ribonucleic acid partially restored the epidermal phenotype of p63-deficient mice. These data provide a novel mechanism by which Satb1, a direct downstream target of p63, contributes in epidermal morphogenesis via establishing tissue-specific chromatin organization and gene expression in epidermal progenitor cells.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/metabolism , Epidermis/embryology , Gene Expression Regulation, Developmental , Matrix Attachment Region Binding Proteins/genetics , Phosphoproteins/genetics , Trans-Activators/genetics , Animals , Cell Differentiation , Epidermal Cells , Genome , In Situ Hybridization, Fluorescence , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphoproteins/metabolism , Trans-Activators/metabolism
7.
Am J Pathol ; 175(3): 1303-14, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700758

ABSTRACT

Bone morphogenetic proteins (BMPs) play pivotal roles in the regulation of skin development. To study the role of BMPs in skin tumorigenesis, BMP antagonist noggin was used to generate keratin 14-targeted transgenic mice. In contrast to wild-type mice, transgenic mice developed spontaneous hair follicle-derived tumors, which resemble human trichofolliculoma. Global gene expression profiles revealed that in contrast to anagen hair follicles of wild-type mice, tumors of transgenic mice showed stage-dependent increases in the expression of genes encoding the selected components of Wnt and Shh pathways. Specifically, expression of the Wnt ligands increased at the initiation stage of tumor formation, whereas expression of the Wnt antagonist and tumor suppressor Wnt inhibitory factor-1 decreased, as compared with fully developed tumors. In contrast, expression of the components of Shh pathway increased in fully developed tumors, as compared with the tumor placodes. Consistent with the expression data, pharmacological treatment of transgenic mice with Wnt and Shh antagonists resulted in the stage-dependent inhibition of tumor initiation, and progression, respectively. Furthermore, BMP signaling stimulated Wnt inhibitory factor-1 expression and promoter activity in cultured tumor cells and HaCaT keratinocytes, as well as inhibited Shh expression, as compared with the corresponding controls. Thus, tumor suppressor activity of the BMPs in skin epithelium depends on the local concentrations of noggin and is mediated at least in part via stage-dependent antagonizing of Wnt and Shh signaling pathways.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Skin Neoplasms/metabolism , Adult , Aged , Animals , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/biosynthesis , Cell Transformation, Neoplastic , Female , Hair Follicle/metabolism , Hair Follicle/pathology , Hedgehog Proteins/metabolism , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Signal Transduction/genetics , Skin/metabolism , Skin/pathology , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Wnt Proteins/metabolism
8.
J Invest Dermatol ; 127(6): 1489-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17273166

ABSTRACT

Alopecia areata (AA) is an autoimmune disorder of the hair follicle characterized by inflammatory cell infiltrates around actively growing (anagen) hair follicles. Substance P (SP) plays a critical role in the cutaneous neuroimmune network and influences immune cell functions through the neurokinin-1 receptor (NK-1R). To better understand the role of SP as an immunomodulatory neuropeptide in AA, we studied its expression and effects on immune cells in a C3H/HeJ mouse model for AA. During early stages of AA development, the number of SP-immunoreactive nerve fibers in skin is increased, compared to non-affected mice. However, during advanced stages of AA, the number of SP-immunoreactive nerves and SP protein levels in skin are decreased, whereas the expression of the SP-degrading enzyme neutral endopeptidase (NEP) is increased, compared to control skin. In AA, NK-1R is expressed on CD8+ lymphocytes and macrophages accumulating around affected hair follicles. Additional SP supply to the skin of AA-affected mice leads to a significant increase of mast cell degranulation and to accelerated hair follicle regression (catagen), accompanied by an increase of CD8+ cells-expressing granzyme B. These data suggest that SP, NEP, and NK-1R serve as important regulators in the molecular signaling network modulating inflammatory response in autoimmune hair loss.


Subject(s)
Alopecia Areata/immunology , Autoimmune Diseases/immunology , Hair Follicle/immunology , Immunologic Factors/immunology , Substance P/immunology , Alopecia Areata/drug therapy , Alopecia Areata/pathology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Degranulation/drug effects , Cell Degranulation/immunology , Disease Models, Animal , Gene Expression/immunology , Granzymes/metabolism , Hair Follicle/innervation , Hair Follicle/pathology , Immunologic Factors/pharmacology , Mast Cells/immunology , Mice , Mice, Inbred C3H , Neprilysin/genetics , Nerve Fibers/immunology , Receptors, Neurokinin-1/metabolism , Signal Transduction/immunology , Substance P/pharmacology
9.
Am J Pathol ; 169(6): 2075-84, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17148670

ABSTRACT

Ectodysplasin (Eda) and its receptor (Edar) are required for normal development of several ectodermal derivatives including hair follicles (HFs). Here, we show that during the murine hair cycle the expression of Eda A1, Edar, Edaradd, and TRAF6 transcripts are minimal in the resting phase and maximal during HF transition from active growth to regression (catagen). Eda A1 mRNA and Edar proteins were expressed in the hair matrix and outer and inner root sheaths of anagen HFs. During catagen, Eda A1 mRNA and Edar protein were expressed in the outer and inner root sheaths and later in the secondary hair germ. Catagen development accompanied by increased apoptosis in the outer root sheath was significantly accelerated in downless mice or after treatment of wild-type mice by a fusion protein that inhibits Edar signaling, compared with the corresponding controls. Microarray, real-time polymerase chain reaction, and immunohistochemical analyses of skin of downless mice revealed a strong decrease of expression of X-linked inhibitor of apoptosis protein (XIAP), compared with the controls, suggesting XIAP as a target for Edar signaling. Thus, our data demonstrate that in addition to its well-established role in HF morphogenesis, Edar signaling is also involved in hair cycle control and regulates apoptosis in HF keratinocytes during catagen.


Subject(s)
Edar Receptor/metabolism , Hair Follicle/growth & development , Hair Follicle/metabolism , RNA, Messenger/metabolism , Signal Transduction , Animals , Ectodysplasins/metabolism , Edar Receptor/genetics , Hair Follicle/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Skin/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
10.
Proc Natl Acad Sci U S A ; 103(48): 18166-71, 2006 Nov 28.
Article in English | MEDLINE | ID: mdl-17114283

ABSTRACT

Bone morphogenetic protein (BMP) signaling is involved in the regulation of a large variety of developmental programs, including those controlling organ sizes. Here, we show that transgenic (TG) mice overexpressing the BMP antagonist noggin (promoter, K5) are characterized by a marked increase in size of anagen hair follicles (HFs) and by the replacement of zig-zag and auchen hairs by awl-like hairs, compared with the age-matched WT controls. Markedly enlarged anagen HFs of TG mice show increased proliferation in the matrix and an increased number of hair cortex and medulla cells compared with WT HFs. Microarray and real-time PCR analyses of the laser-captured hair matrix cells show a strong decrease in expression of Cdk inhibitor p27(Kip1) and increased expression of selected cyclins in TG vs. WT mice. Similar to TG mice, p27(Kip1) knockout mice also show an increased size of anagen HFs associated with increased cell proliferation in the hair bulb. Primary epidermal keratinocytes (KC) from TG mice exhibit significantly increased proliferation and decreased p27(Kip1) expression, compared with WT KC. Alternatively, activation of BMP signaling in HaCaT KC induces growth arrest, stimulates p27(Kip1) expression, and positively regulates p27(Kip1) promoter activity, thus further supporting a role of p27(Kip1) in mediating the effects of BMP signaling on HF size. These data suggest that BMP signaling plays an important role in regulating cell proliferation and controls the size of anagen HFs by modulating the expression of cell-cycle-associated genes in hair matrix KC.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Hair Follicle/cytology , Hair Follicle/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hair/cytology , Hair/metabolism , Hair Follicle/growth & development , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Transgenic
11.
J Invest Dermatol ; 125(6): 1259-67, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354197

ABSTRACT

Melanin synthesis in the hair follicle (HF) is strictly coupled to the growth stage of the hair cycle and is interrupted during follicle regression (catagen) and resting. Using tyrosine-related protein 2 (Trp)2-LacZ transgenic mice as a model, we show that distinct melanocyte subpopulations of the HF display distinct patterns of apoptosis and survival during catagen. Melanocytes located in the outer root sheath express Bcl-2 and are TUNEL-negative. Part of the pigment-producing melanocytes located above the follicular papilla expresses Fas, TUNEL, and is likely to undergo apoptosis, whereas the other part of these melanocytes expresses c-kit, Bcl-2, and becomes visible in the follicular papilla. During late catagen, TUNEL and Ki-67 negative melanocytes expressing Bcl-2 are seen in the secondary germ of the HF. Lack of proliferation in the follicular melanocytes during catagen suggests that secondary hair germ of late catagen HF is most likely repopulated by melanocytes arising from the outer root sheath or follicular papilla of early/mid-catagen HF. Taken together, these data suggest a possible scenario and mechanisms of the remodeling of the follicular pigmentary unit during HF anagen-catagen-telogen transition and may be used for the establishing in vivo models for pharmacological modulation of melanocyte apoptosis and survival during the hair cycle.


Subject(s)
Hair Follicle/cytology , Hair Follicle/physiology , Melanocytes/cytology , Animals , Apoptosis , In Situ Nick-End Labeling , Melanocytes/physiology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Models, Animal , Plant Roots/cytology , Plant Roots/physiology , beta-Galactosidase/genetics
12.
Proc Natl Acad Sci U S A ; 102(1): 93-8, 2005 Jan 04.
Article in English | MEDLINE | ID: mdl-15618398

ABSTRACT

Hair pigmentation is controlled by tightly coordinated programs of melanin synthesis and involves signaling through the melanocortin type 1 receptor (MC-1R) that regulates the switch between pheomelanogenesis and eumelanogenesis. However, the involvement of other signaling systems, including the bone morphogenetic protein (BMP) pathway, in the control of hair pigmentation remains to be elucidated. To assess the effects of BMP signaling on hair pigmentation, transgenic mice overexpressing the BMP antagonist noggin (promoter: keratin 5) were generated. Whereas wild-type C3H/HeJ mice have a subapical yellow band on otherwise black dorsal hairs, K5-Noggin mice are characterized by the absence of a yellow band and near-black pigment in dorsal coat. Noggin overexpression is accompanied by strongly reduced levels of Agouti signal protein and enhanced expression of microphthalmia transcription factor in the midphase of the hair-growth cycle. Wild-type color in K5-Noggin mice is restored by administration of a synthetic MC-1R antagonist resulting in the reappearance of a subapical yellow band. BMP-4 stimulates the expression of Agouti transcripts and protein in primary epidermal keratinocytes, and BMP signaling positively regulates dermal papilla-specific enhancer of the Agouti gene in primary dermal fibroblasts. Taken together, these data suggests that BMP signaling controls the expression of Agouti protein in the hair follicle and provide evidence for interaction between BMP and MC-1R signaling pathways to modulate the balance between pheomelanogenesis and eumelanogenesis during hair growth.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Hair/metabolism , Pigmentation/physiology , Receptor, Melanocortin, Type 1/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Morphogenetic Protein Receptors , Carrier Proteins , DNA-Binding Proteins/metabolism , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor , Proteins/genetics , Proteins/metabolism , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Smad Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism
13.
Cancer Res ; 64(17): 6266-70, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15342414

ABSTRACT

Chemotherapeutic agents induce p53-dependent apoptosis in the hair follicle (HF) resulting in hair loss, a common side effect of cancer therapy. Here, we show that Fas as a p53 target plays important role in the HF response to cyclophosphamide. Specifically, we demonstrate that Fas is up-regulated in HF keratinocytes after cyclophosphamide treatment, Fas ligand-neutralizing antibody partially inhibits HF response to cyclophosphamide in wild-type mice, and Fas knockout mice show significant retardation of cyclophosphamide-induced HF involution associated with reduced Fas-associated death domain and caspase-8 expression. These data raise a possibility to explore blockade of Fas signaling as a part of complex local therapy for inhibiting keratinocyte apoptosis and hair loss induced by chemotherapy.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents, Alkylating/pharmacology , Cyclophosphamide/pharmacology , Hair Follicle/drug effects , Hair Follicle/physiology , fas Receptor/physiology , Animals , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Carrier Proteins/biosynthesis , Caspase 8 , Caspases/biosynthesis , Fas Ligand Protein , Fas-Associated Death Domain Protein , Female , In Situ Nick-End Labeling , Keratinocytes/cytology , Keratinocytes/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , fas Receptor/metabolism
14.
EMBO J ; 22(12): 2992-3003, 2003 Jun 16.
Article in English | MEDLINE | ID: mdl-12805214

ABSTRACT

Contact of developing sensory organs with the external environment is established via the formation of openings in the skin. During eye development, eyelids first grow, fuse and finally reopen, thus providing access for visual information to the retina. Here, we show that eyelid opening is strongly inhibited in transgenic mice overexpressing the bone morphogenetic protein (BMP) antagonist noggin from the keratin 5 (K5) promoter in the epidermis. In wild-type mice, enhanced expression of the kinase-inactive form of BMPR-IB mediated by an adenovirus vector also inhibits eyelid opening. Noggin overexpression leads to reduction of apoptosis and retardation of cell differentiation in the eyelid epithelium, which is associated with downregulation of expression of the apoptotic receptors (Fas, p55 kDa TNFR), Id3 protein and keratinocyte differentiation markers (loricrin, involucrin). BMP-4, but not EGF or TGF-alpha, accelerates opening of the eyelid explants isolated from K5-Noggin transgenic mice when cultured ex vivo. These data suggest that the BMP signaling pathway plays an important role in regulation of genetic programs of eyelid opening and skin remodeling during the final steps of eye morphogenesis.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Epidermal Cells , Eyelids/growth & development , Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Biomarkers , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Carrier Proteins , Culture Techniques , DNA-Binding Proteins/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Epidermal Growth Factor/metabolism , Epidermis/growth & development , Epidermis/physiology , Eyelids/cytology , Genetic Vectors , Growth Differentiation Factor 5 , Humans , In Situ Hybridization , In Situ Nick-End Labeling , Keratin-15 , Keratin-5 , Keratinocytes/cytology , Keratinocytes/physiology , Keratins/genetics , Mice , Mice, Transgenic , Morphogenesis/physiology , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Proteins/genetics , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Signal Transduction/physiology , Smad Proteins , Trans-Activators/metabolism , Transforming Growth Factor alpha/metabolism
15.
J Invest Dermatol ; 120(1): 27-35, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12535195

ABSTRACT

Chemotherapy alters the structure and function of hair follicle melanocytes. Molecular mechanisms controlling melanocyte responses during chemotherapy-induced hair loss, however, remain largely unknown. Using immunohistology and multicolor confocal microscopy, we show here that cyclophosphamide administration to C57BL/6 mice alters the activity and fate of hair follicle melanocytes. After 24-48 h, hair bulb melanocytes expressing Fas undergo apoptosis. The number of apoptotic follicular melanocytes is significantly reduced (p<0.01) in cyclophosphamide-treated Fas knockout mice compared to wild-type controls, suggesting that Fas signaling contributes to chemotherapy-induced melanocyte death. After 3-5 d, surviving hair bulb melanocytes express c-kit receptor, proliferate, and appear to migrate up the outer root sheath. Tyrosinase-positive and melanogenically active cells then appear in the epidermis. By Western blotting and immunohistochemistry, expression levels of the c-kit ligand, stem cell factor, in skin and epidermis are strongly increased after cyclophosphamide treatment. Cyclophosphamide-induced migration of the hair follicle melanocytes into epidermis is completely abrogated by administration of c-kit neutralizing antibody. These data suggest that chemotherapy induces a complex response in the hair follicle melanocytes, which includes apoptosis, proliferation, and migration. Pharmacologic manipulation of Fas and c-kit signaling pathways might be useful for the correction of skin hyperpigmentation as a side-effect of chemotherapy.


Subject(s)
Alopecia/chemically induced , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cyclophosphamide/toxicity , Hair Follicle/drug effects , Melanocytes/drug effects , Proto-Oncogene Proteins c-kit/physiology , fas Receptor/physiology , Alopecia/pathology , Animals , Cell Division/drug effects , Cell Movement/drug effects , Female , Hair Follicle/pathology , Melanocytes/pathology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...