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










Publication year range
1.
J Invest Dermatol ; 138(2): 256-264, 2018 02.
Article in English | MEDLINE | ID: mdl-28942365

ABSTRACT

The transcription factor NF-κB controls key features of hair follicle (HF) development, but the role of NF-κB in adult HF cycle regulation remains obscure. Using NF-κB reporter mouse models, strong NF-κB activity was detected in the secondary hair germ of late telogen and early anagen HFs, suggesting a potential role for NF-κB in HF stem/progenitor cell activation during anagen induction. At mid-anagen, NF-κB activity was observed in the inner root sheath and unilaterally clustered in the HF matrix, which indicates that NF-κB activity is also involved in hair fiber morphogenesis during HF cycling. A mouse model with inducible NF-κB suppression in the epithelium revealed pelage hair-type-dependent functions of NF-κB in cycling HFs. NF-κB participates in telogen-anagen transition in awl and zigzag HFs, and is required for zigzag hair bending and guard HF cycling. Interestingly, zigzag hair shaft bending depends on noncanonical NF-κB signaling, which previously has only been associated with lymphoid cell biology. Furthermore, loss of guard HF cycling suggests that in this particular hair type, NF-κB is indispensable for stem cell activation, maintenance, and/or growth.


Subject(s)
Hair Follicle/growth & development , Morphogenesis/physiology , NF-kappa B/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Gene Expression Regulation , Mice , Mice, Transgenic , Models, Animal , NF-kappa B/genetics
2.
Matrix Biol ; 56: 42-56, 2016 12.
Article in English | MEDLINE | ID: mdl-27234308

ABSTRACT

Laminins are the most abundant non-collagenous basement membrane (BM) components, composed of an α, ß and γ chain. The laminin γ1 chain, encoded by LAMC1, is the most abundant γ chain. The main laminin isoforms in the dermo-epidermal junction (DEJ) are laminin-332, laminin-511 and laminin-211, the latter being restricted to the lower part of hair follicles (HFs). Complete deletion of LAMC1 results in lethality around embryonic day 5.5. To study the function of laminin γ1 containing isoforms in skin development and maturation after birth, we generated mice lacking LAMC1 expression in basal keratinocytes (LAMC1EKO) using the keratin 14 (K14) Cre/loxP system. This deletion resulted in loss of keratinocyte derived laminin-511 and in deposition of fibroblast derived laminin-211 throughout the whole DEJ. The DEJ in areas between hemidesmosomes was thickened, whereas hemidesmosome morphology was normal. Most strikingly, LAMC1EKO mice showed delayed HF morphogenesis accompanied by reduced proliferation of hair matrix cells and impaired differentiation of hair shafts (HS). However, this deletion did not interfere with early HF development, since placode numbers and embryonic hair germ formation were not affected. Microarray analysis of skin revealed down regulation of mainly different hair keratins. This is due to reduced expression of transcription factors such as HoxC13, FoxN1, FoxQ1 and Msx2, known to regulate expression of hair keratins. While the role of laminin-511 in signaling during early hair germ formation and elongation phase has been described, we here demonstrate that epidermal laminin-511 is also a key regulator for later hair development and HS differentiation.


Subject(s)
Hair Follicle/growth & development , Laminin/genetics , Animals , Basement Membrane/metabolism , Cell Differentiation , Cells, Cultured , Gene Deletion , Gene Expression , Hair Follicle/cytology , Hair Follicle/embryology , Keratinocytes/metabolism , Laminin/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis
4.
J Invest Dermatol ; 136(1): 34-44, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26763421

ABSTRACT

Hair follicles (HFs) undergo lifelong cyclical transformations, progressing through stages of rapid growth (anagen), regression (catagen), and relative "quiescence" (telogen). Given that HF cycling abnormalities underlie many human hair growth disorders, the accurate classification of individual cycle stages within skin biopsies is clinically important and essential for hair research. For preclinical human hair research purposes, human scalp skin can be xenografted onto immunocompromised mice to study human HF cycling and manipulate long-lasting anagen in vivo. Although available for mice, a comprehensive guide on how to recognize different human hair cycle stages in vivo is lacking. In this article, we present such a guide, which uses objective, well-defined, and reproducible criteria, and integrates simple morphological indicators with advanced, (immuno)-histochemical markers. This guide also characterizes human HF cycling in xenografts and highlights the utility of this model for in vivo hair research. Detailed schematic drawings and representative micrographs provide examples of how best to identify human HF stages, even in suboptimally sectioned tissue, and practical recommendations are given for designing human-on-mouse hair cycle experiments. Thus, this guide seeks to offer a benchmark for human hair cycle stage classification, for both hair research experts and newcomers to the field.


Subject(s)
Cell Cycle/physiology , Hair Follicle/growth & development , Hair/physiology , Animals , Apoptosis/physiology , Biopsy, Needle , Cells, Cultured , Guidelines as Topic , Hair Follicle/anatomy & histology , Heterografts , Humans , Immunohistochemistry , Mice , Mice, SCID
5.
Exp Dermatol ; 24(12): 903-11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26284830

ABSTRACT

For almost a quarter of a century, ex vivo studies of human scalp hair follicles (HFs) have permitted major advances in hair research, spanning diverse fields such as chronobiology, endocrinology, immunology, metabolism, mitochondrial biology, neurobiology, pharmacology, pigmentation and stem cell biology. Despite this, a comprehensive methodological guide to serum-free human HF organ culture (HFOC) that facilitates the selection and analysis of standard HF biological parameters and points out both research opportunities and pitfalls to newcomers to the field is still lacking. The current methods review aims to close an important gap in the literature and attempts to promote standardisation of human HFOC. We provide basic information outlining the establishment of HFOC through to detailed descriptions of the analysis of standard read-out parameters alongside practical examples. The guide closes by pointing out how serum-free HFOC can be utilised optimally to obtain previously inaccessible insights into human HF biology and pathology that are of interest to experimental dermatologists, geneticists, developmental biologists and (neuro-) endocrinologists alike and by highlighting novel applications of the model, including gene silencing and gene expression profiling of defined, laser capture-microdissected HF compartments.


Subject(s)
Hair Follicle/growth & development , Organ Culture Techniques/methods , Apoptosis , Cell Proliferation , Culture Media, Serum-Free , Hair Color , Hair Follicle/anatomy & histology , Hair Follicle/physiology , Humans , Keratinocytes/cytology , Organ Culture Techniques/trends
6.
Arch Dermatol Res ; 307(9): 841-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26216444

ABSTRACT

Spermidine (Spd), the prototypic polyamine, has been shown to be essential for hair follicle (HF) growth. However, Spd can be readily converted into other polyamines, and is physiologically unstable. Therefore, to assess its individual functions on HFs, we used the metabolically stable Spd analog N(1)-methylspermidine (N(1)-MeSpd). N(1)-MeSpd was confirmed to be a metabolically stable compound, with a half life of 90 h. 0.5 µM N(1)-MeSpd strongly prolonged anagen and decreased cell apoptosis in HFs in culture after 6 days, accompanied by specific stimulation of the expression of the epithelial stem cell-associated keratin, K15. N(1)-MeSpd also reduced lactate dehydrogenase activity in the culture supernatant, a parameter of cell death and cell lysis. N(1)-MeSpd diminished intracellular reactive oxygen species production in cultured keratinocytes, and reduced tumor necrosis factor-α, interleukin (IL)-1ß and IL-6 gene and protein expression after lipopolysaccharide stimulation. This suggests that some effects of N(1)-MeSpd may be mediated by anti-oxidative and anti-inflammatory effects. These additional properties of N(1)-MeSpd could be clinically important for the treatment of inflammatory alopecias and inflammatory scalp diseases.


Subject(s)
Epithelial Cells/metabolism , Hair Follicle/metabolism , Loose Anagen Hair Syndrome/pathology , Spermidine/analogs & derivatives , Spermidine/metabolism , Stem Cells/metabolism , Cell Line , Cell Proliferation , Epithelial Cells/cytology , Hair Follicle/cytology , Humans , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Keratin-15/biosynthesis , Keratin-19/biosynthesis , Keratinocytes/metabolism , L-Lactate Dehydrogenase/metabolism , Loose Anagen Hair Syndrome/genetics , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Stem Cells/cytology , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Invest Dermatol ; 135(3): 679-689, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25371971

ABSTRACT

Here, we studied how epithelial energy metabolism impacts overall skin development by selectively deleting intraepithelial mtDNA in mice by ablating a key maintenance factor (Tfam(EKO)), which induces loss of function of the electron transport chain (ETC). Quantitative (immuno)histomorphometry demonstrated that Tfam(EKO) mice showed significantly reduced hair follicle (HF) density and morphogenesis, fewer intrafollicular keratin15+ epithelial progenitor cells, increased apoptosis, and reduced proliferation. Tfam(EKO) mice also displayed premature entry into (aborted) HF cycling by apoptosis-driven HF regression (catagen). Ultrastructurally, Tfam(EKO) mice exhibited severe HF dystrophy, pigmentary abnormalities, and telogen-like condensed dermal papillae. Epithelial HF progenitor cell differentiation (Plet1, Lrig1 Lef1, and ß-catenin), sebaceous gland development (adipophilin, Scd1, and oil red), and key mediators/markers of epithelial-mesenchymal interactions during skin morphogenesis (NCAM, versican, and alkaline phosphatase) were all severely altered in Tfam(EKO) mice. Moreover, the number of mast cells, major histocompatibility complex class II+, or CD11b+ immunocytes in the skin mesenchyme was increased, and essentially no subcutis developed. Therefore, in contrast to their epidermal counterparts, pilosebaceous unit stem cells depend on a functional ETC. Most importantly, our findings point toward a frontier in skin biology: the coupling of HF keratinocyte mitochondrial function with the epithelial-mesenchymal interactions that drive overall development of the skin and its appendages.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Hair Follicle/growth & development , Mitochondria/physiology , Morphogenesis/physiology , Skin Physiological Phenomena , Animals , Apoptosis/physiology , Cell Proliferation , DNA, Mitochondrial/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Energy Metabolism/physiology , Epithelium/physiology , Hair Follicle/cytology , High Mobility Group Proteins/deficiency , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Mice , Mice, Knockout , Models, Animal
8.
J Clin Invest ; 124(9): 3713-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25061872

ABSTRACT

The endocannabinoid system (ECS) regulates multiple physiological processes, including cutaneous cell growth and differentiation. Here, we explored the effects of the major nonpsychotropic phytocannabinoid of Cannabis sativa, (-)-cannabidiol (CBD), on human sebaceous gland function and determined that CBD behaves as a highly effective sebostatic agent. Administration of CBD to cultured human sebocytes and human skin organ culture inhibited the lipogenic actions of various compounds, including arachidonic acid and a combination of linoleic acid and testosterone, and suppressed sebocyte proliferation via the activation of transient receptor potential vanilloid-4 (TRPV4) ion channels. Activation of TRPV4 interfered with the prolipogenic ERK1/2 MAPK pathway and resulted in the downregulation of nuclear receptor interacting protein-1 (NRIP1), which influences glucose and lipid metabolism, thereby inhibiting sebocyte lipogenesis. CBD also exerted complex antiinflammatory actions that were coupled to A2a adenosine receptor-dependent upregulation of tribbles homolog 3 (TRIB3) and inhibition of the NF-κB signaling. Collectively, our findings suggest that, due to the combined lipostatic, antiproliferative, and antiinflammatory effects, CBD has potential as a promising therapeutic agent for the treatment of acne vulgaris.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabidiol/pharmacology , Sebaceous Glands/drug effects , Acne Vulgaris/drug therapy , Acne Vulgaris/etiology , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Lipogenesis/drug effects , Sebaceous Glands/cytology , Sebaceous Glands/pathology , Sebum/physiology , TRPV Cation Channels/physiology
9.
Mol Cell Biol ; 34(16): 3086-95, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891618

ABSTRACT

The epidermal growth factor receptor (EGFR) system is a key regulator of epithelial development and homeostasis. Its functions in the sebaceous gland (SG), however, remain poorly characterized. In this study, using a transgenic mouse line with tissue-specific and inducible expression of the EGFR ligand epigen, we showed that increased activation of the EGFR in skin keratinocytes results in enlarged SGs and increased sebum production. The phenotype can be reverted by interrupting transgene expression and is EGFR dependent, as gland size and sebum levels return to normal values after crossing to the EGFR-impaired mouse line Wa5. Intriguingly, however, the SG enlargement appears only if EGFR activation occurs before birth. Importantly, the enlarged sebaceous glands are associated with an increased expression of the transcription factor MYC and of the transmembrane proteins LRIG1, an established negative-feedback regulator of the EGFR/ERBB tyrosine kinase receptors and a stem cell marker. Our findings identify EGFR signaling as a major pathway determining SG activity and suggest a functional relationship between the EGFR/ERBB system and MYC/LRIG1 in the commitment of stem cells toward specific progenitor cell types, with implications for our understanding of their role in tissue development, homeostasis, and disease.


Subject(s)
Epidermal Growth Factor/biosynthesis , ErbB Receptors/biosynthesis , Sebaceous Glands/embryology , Sebaceous Glands/pathology , Animals , Epidermal Growth Factor/genetics , Epidermis/growth & development , Epidermis/pathology , Epigen , ErbB Receptors/genetics , Hair Follicle/growth & development , Hair Follicle/pathology , Hyperplasia/metabolism , Keratinocytes/metabolism , Membrane Glycoproteins/biosynthesis , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , Sebaceous Glands/metabolism , Sebum/metabolism , Signal Transduction/genetics
12.
Biol Rev Camb Philos Soc ; 89(3): 618-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24299058

ABSTRACT

For over a century, frogs have been studied across various scientific fields, including physiology, embryology, neuroscience, (neuro)endocrinology, ecology, genetics, behavioural science, evolution, drug development, and conservation biology. In some cases, frog skin has proven very successful as a research model, for example aiding in the study of ion transport through tight epithelia, where it has served as a model for the vertebrate distal renal tubule and mammalian epithelia. However, it has rarely been considered in comparative studies involving human skin. Yet, despite certain notable adaptations that have enabled frogs to survive in both aquatic and terrestrial environments, frog skin has many features in common with human skin. Here we present a comprehensive overview of frog (and toad) skin ontogeny, anatomy, cytology, neuroendocrinology and immunology, with special attention to its unique adaptations as well as to its similarities with the mammalian integument, including human skin. We hope to provide a valuable reference point and a source of inspiration for both amphibian investigators and mammalian researchers studying the structural and functional properties of the largest organ of the vertebrate body.


Subject(s)
Anura/physiology , Skin Physiological Phenomena , Animals , Humans
13.
J Invest Dermatol ; 134(3): 610-619, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24005054

ABSTRACT

The hair follicle (HF) is a continuously remodeled mini organ that cycles between growth (anagen), regression (catagen), and relative quiescence (telogen). As the anagen-to-catagen transformation of microdissected human scalp HFs can be observed in organ culture, it permits the study of the unknown controls of autonomous, rhythmic tissue remodeling of the HF, which intersects developmental, chronobiological, and growth-regulatory mechanisms. The hypothesis that the peripheral clock system is involved in hair cycle control, i.e., the anagen-to-catagen transformation, was tested. Here we show that in the absence of central clock influences, isolated, organ-cultured human HFs show circadian changes in the gene and protein expression of core clock genes (CLOCK, BMAL1, and Period1) and clock-controlled genes (c-Myc, NR1D1, and CDKN1A), with Period1 expression being hair cycle dependent. Knockdown of either BMAL1 or Period1 in human anagen HFs significantly prolonged anagen. This provides evidence that peripheral core clock genes modulate human HF cycling and are an integral component of the human hair cycle clock. Specifically, our study identifies BMAL1 and Period1 as potential therapeutic targets for modulating human hair growth.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Rhythm/physiology , Hair Follicle/physiology , Period Circadian Proteins/genetics , Scalp/physiology , ARNTL Transcription Factors/metabolism , Adult , Aged , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation/physiology , Gene Silencing , Hair Follicle/cytology , Hair Follicle/growth & development , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Male , Middle Aged , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Organ Culture Techniques , Period Circadian Proteins/metabolism , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Scalp/cytology , Scalp/growth & development
14.
Exp Dermatol ; 22(10): 631-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24079731

ABSTRACT

The skin of most mammals is characterised by the presence of sebaceous glands (SGs), whose predominant constituent cell population is sebocytes, that is, lipid-producing epithelial cells, which develop from the hair follicle. Besides holocrine sebum production (which contributes 90% of skin surface lipids), multiple additional SG functions have emerged. These range from antimicrobial peptide production and immunomodulation, via lipid and hormone synthesis/metabolism, to the provision of an epithelial progenitor cell reservoir. Therefore, in addition to its involvement in common skin diseases (e.g. acne vulgaris), the unfolding diversity of SG functions, both in skin health and disease, has raised interest in this integral component of the pilosebaceous unit. This practical guide provides an introduction to SG biology and to relevant SG histochemical and immunohistochemical techniques, with emphasis placed on in situ evaluation methods that can be easily employed. We propose a range of simple, established markers, which are particularly instructive when addressing specific SG research questions in the two most commonly investigated species in SG research, humans and mice. To facilitate the development of reproducible analysis techniques for the in situ evaluation of SGs, this methods review concludes by suggesting quantitative (immuno-)histomorphometric methods for standardised SG evaluation.


Subject(s)
Sebaceous Glands/physiology , Skin/pathology , Acne Vulgaris/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Apoptosis , Cell Proliferation , Dermatology/methods , Epithelial Cells/metabolism , Hair Follicle/metabolism , Humans , Immunohistochemistry , Lipids/biosynthesis , Mice , Microscopy, Fluorescence , Sebaceous Glands/anatomy & histology , Sebum/metabolism , Skin/metabolism , Skin Diseases/metabolism
15.
PLoS One ; 8(9): e73596, 2013.
Article in English | MEDLINE | ID: mdl-24023889

ABSTRACT

There remains a critical need for new therapeutics that promote wound healing in patients suffering from chronic skin wounds. This is, in part, due to a shortage of simple, physiologically and clinically relevant test systems for investigating candidate agents. The skin of amphibians possesses a remarkable regenerative capacity, which remains insufficiently explored for clinical purposes. Combining comparative biology with a translational medicine approach, we report the development and application of a simple ex vivo frog (Xenopus tropicalis) skin organ culture system that permits exploration of the effects of amphibian skin-derived agents on re-epithelialisation in both frog and human skin. Using this amphibian model, we identify thyrotropin-releasing hormone (TRH) as a novel stimulant of epidermal regeneration. Moving to a complementary human ex vivo wounded skin assay, we demonstrate that the effects of TRH are conserved across the amphibian-mammalian divide: TRH stimulates wound closure and formation of neo-epidermis in organ-cultured human skin, accompanied by increased keratinocyte proliferation and wound healing-associated differentiation (cytokeratin 6 expression). Thus, TRH represents a novel, clinically relevant neuroendocrine wound repair promoter that deserves further exploration. These complementary frog and human skin ex vivo assays encourage a comparative biology approach in future wound healing research so as to facilitate the rapid identification and preclinical testing of novel, evolutionarily conserved, and clinically relevant wound healing promoters.


Subject(s)
Re-Epithelialization/drug effects , Skin/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Aged , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Estrogens/pharmacology , Evolution, Molecular , Female , Humans , Keratin-6/metabolism , Male , Middle Aged , Protein Precursors/metabolism , Serum/metabolism , Skin/cytology , Skin/metabolism , Up-Regulation/drug effects , Xenopus
16.
J Pathol ; 231(2): 236-47, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23788005

ABSTRACT

Lichen planopilaris (LPP) is a chronic inflammatory disease of unknown pathogenesis that leads to permanent hair loss. Whilst destruction of epithelial hair follicle stem cells (eHFSCs) that reside in an immunologically protected niche of the HF epithelium, the bulge, is a likely key event in LPP pathogenesis, this remains to be demonstrated. We have tested the hypotheses that bulge immune privilege (IP) collapse and inflammation-induced eHFSC death are key components in the pathogenesis of LPP. Biopsies of lesional and non-lesional scalp skin from adult LPP patients (n = 42) were analysed by quantitative (immuno)histomorphometry, real-time quantitative polymerase chain reaction (qRT-PCR), laser capture microdissection and microarray analysis, or skin organ culture. At both the protein and transcriptional level, lesional LPP HFs showed evidence for bulge IP collapse (ie increased expression of MHC class I and II, ß2microglobulin; reduced TGFß2 and CD200 expression). This was accompanied by a Th1-biased cytotoxic T cell response (ie increased CD8(+) GranzymeB(+) T cells and CD123(+) plasmacytoid dendritic cells, with increased CXCR3 expression) and increased expression of interferon-inducible chemokines (CXCL9/10/11). Interestingly, lesional LPP eHFSCs showed both increased proliferation and apoptosis in situ. Microarray analysis revealed a loss of eHFSC signatures and increased expression of T cell activation/binding markers in active LPP, while bulge PPARγ transcription was unaltered compared to non-lesional LPP HFs. In organ culture of non-lesional LPP skin, interferon-γ (IFNγ) induced bulge IP collapse. LPP is an excellent model disease for studying and preventing immune destruction of human epithelial stem cells in situ. These novel findings raise the possibility that LPP represents an autoimmune disease in whose pathogenesis IFNγ-induced bulge IP collapse plays an important role. Therapeutically, bulge IP protection/restoration may help to better manage this highly treatment-resistant cicatricial alopecia.


Subject(s)
Alopecia/pathology , Hair Follicle/pathology , Lichen Planus/pathology , Stem Cell Niche , Alopecia/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Hair Follicle/immunology , Humans , Immunohistochemistry , Laser Capture Microdissection , Lichen Planus/immunology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Stem Cells/immunology , Stem Cells/pathology
17.
J Allergy Clin Immunol ; 132(1): 182-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23453134

ABSTRACT

BACKGROUND: Because many chronic inflammatory and allergic disorders are intimately linked to excessive mast cell (MC) numbers and activation, it is clinically important to understand the physiologic mechanisms preventing excess MC accumulation/degranulation in normal human tissues. OBJECTIVE: Because endocannabinoids are increasingly recognized as neuroendocrine regulators of MC biology, we investigated how cannabinoid receptor (CB) 1 signaling affects human mucosal-type mast cells (hMMCs). METHODS: Using organ-cultured nasal polyps as a surrogate tissue for human bronchial mucosa, we investigated how CB1 stimulation, inhibition, or knockdown affects hMMC biology using quantitative (immuno)histomorphometry and electron microscopy. RESULTS: Kit(+) hMMCs express functional CB1 in situ. Blockade of CB1 signaling (with the specific CB1 antagonist N-(piperidin-1-yl)-1-(2,4-dichlorophenyl)-5-(4-chlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide [AM251] or CB1 gene knockdown) enhanced hMMC degranulation and increased total numbers without affecting their proliferation in situ. This suggests that inhibiting CB1 signaling induces hMMC maturation from resident progenitor cells within human mucosal stroma. hMMC maturation was induced at least in part through upregulating stem cell factor production. Both the prototypic endocannabinoid anandamide and the CB1-selective agonist arachidonyl-2-chloroethylamide effectively counteracted secretagogue-triggered excessive hMMC degranulation. CONCLUSIONS: The current serum-free nasal polyp organ culture model allows physiologically and clinically relevant insights into the biology and pharmacologic responses of primary hMMCs in situ. In human airway mucosa hMMC activation and maturation are subject to a potent inhibitory endocannabinoid tone through CB1 stimulation. This invites one to target the endocannabinoid system in human airway mucosa as a novel strategy in the future management of allergic diseases.


Subject(s)
Cell Degranulation , Mast Cells/physiology , Receptor, Cannabinoid, CB1/physiology , Adult , Aged , Aged, 80 and over , Cell Movement , Endocannabinoids/pharmacology , Female , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-kit/analysis , Stem Cell Factor/biosynthesis
18.
J Invest Dermatol ; 133(6): 1591-600, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23334344

ABSTRACT

P-cadherin serves as a major topobiological cue in mammalian epithelium. In human hair follicles (HFs), it is prominently expressed in the inner hair matrix that harbors the HF pigmentary unit. However, the role of P-cadherin in normal human pigmentation remains unknown. As patients with mutations in the gene that encodes P-cadherin show hypotrichosis and fair hair, we explored the hypothesis that P-cadherin may control HF pigmentation. When P-cadherin was silenced in melanogenically active organ-cultured human scalp HFs, this significantly reduced HF melanogenesis and tyrosinase activity as well as gene and/or protein expression of gp100, stem cell factor, c-Kit, and microphthalmia-associated transcription factor (MITF), both in situ and in isolated human HF melanocytes. Instead, epidermal pigmentation was unaffected by P-cadherin knockdown in organ-cultured human skin. In hair matrix keratinocytes, P-cadherin silencing reduced plasma membrane ß-catenin, whereas glycogen synthase kinase 3 beta (GSK3ß) and phospho-ß-catenin expression were significantly upregulated. This suggests that P-cadherin-GSK3ß/Wnt signaling is required for maintaining the expression of MITF to sustain intrafollicular melanogenesis. Thus, P-cadherin-mediated signaling is a melanocyte subtype-specific topobiological regulator of normal human pigmentation, possibly via GSK3ß-mediated canonical Wnt signaling.


Subject(s)
Cadherins/metabolism , Hair Follicle/cytology , Hair Follicle/physiology , Melanocytes/cytology , Melanocytes/physiology , Skin Pigmentation/physiology , Cadherins/genetics , Cells, Cultured , Enzyme Activation/physiology , Epidermal Cells , Epidermis/physiology , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Melanins/biosynthesis , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Wnt Signaling Pathway/physiology , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
20.
PLoS One ; 8(12): e84356, 2013.
Article in English | MEDLINE | ID: mdl-24386370

ABSTRACT

ß1 integrin regulates multiple epithelial cell functions by connecting cells with the extracellular matrix (ECM). While ß1 integrin-mediated signaling in murine epithelial stem cells is well-studied, its role in human adult epithelial progenitor cells (ePCs) in situ remains to be defined. Using microdissected, organ-cultured human scalp hair follicles (HFs) as a clinically relevant model for studying human ePCs within their natural topobiological habitat, ß1 integrin-mediated signaling in ePC biology was explored by ß1 integrin siRNA silencing, specific ß1 integrin-binding antibodies and pharmacological inhibition of integrin-linked kinase (ILK), a key component of the integrin-induced signaling cascade. ß1 integrin knock down reduced keratin 15 (K15) expression as well as the proliferation of outer root sheath keratinocytes (ORSKs). Embedding of HF epithelium into an ECM rich in ß1 integrin ligands that mimic the HF mesenchyme significantly enhanced proliferation and migration of ORSKs, while K15 and CD200 gene and protein expression were inhibited. Employing ECM-embedded ß1 integrin-activating or -inhibiting antibodies allowed to identify functionally distinct human ePC subpopulations in different compartments of the HF epithelium. The ß1 integrin-inhibitory antibody reduced ß1 integrin expression in situ and selectively enhanced proliferation of bulge ePCs, while the ß1 integrin-stimulating antibody decreased hair matrix keratinocyte apoptosis and enhanced transferrin receptor (CD71) immunoreactivity, a marker of transit amplifying cells, but did not affect bulge ePC proliferation. That the putative ILK inhibitor QLT0267 significantly reduced ORSK migration and proliferation and induced massive ORSK apoptosis suggests a key role for ILK in mediating the ß1 integrin effects. Taken together, these findings demonstrate that ePCs in human HFs require ß1 integrin-mediated signaling for survival, adhesion, and migration, and that different human HF ePC subpopulations differ in their response to ß1 integrin signaling. These insights may be exploited for cell-based regenerative medicine strategies that employ human HF-derived ePCs.


Subject(s)
Apoptosis , Cell Movement , Epithelial Cells/cytology , Integrin beta1/metabolism , Signal Transduction , Stem Cells/cytology , Adult , Aged , Apoptosis/drug effects , Azo Compounds/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival , DNA/biosynthesis , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Gene Silencing , Hair Follicle/cytology , Humans , Integrin beta1/genetics , Keratinocytes/cytology , Keratinocytes/drug effects , Ligands , Middle Aged , Pyrazoles/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...