Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 1029818, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439142

RESUMO

This review presents several aspects of the innovative concept of sebaceous immunobiology, which summarizes the numerous activities of the sebaceous gland including its classical physiological and pathophysiological tasks, namely sebum production and the development of seborrhea and acne. Sebaceous lipids, which represent 90% of the skin surface lipids in adolescents and adults, are markedly involved in the skin barrier function and perifollicular and dermal innate immune processes, leading to inflammatory skin diseases. Innovative experimental techniques using stem cell and sebocyte models have clarified the roles of distinct stem cells in sebaceous gland physiology and sebocyte function control mechanisms. The sebaceous gland represents an integral part of the pilosebaceous unit and its status is connected to hair follicle morphogenesis. Interestingly, professional inflammatory cells contribute to sebocyte differentiation and homeostasis, whereas the regulation of sebaceous gland function by immune cells is antigen-independent. Inflammation is involved in the very earliest differentiation changes of the pilosebaceous unit in acne. Sebocytes behave as potent immune regulators, integrating into the innate immune responses of the skin. Expressing inflammatory mediators, sebocytes also contribute to the polarization of cutaneous T cells towards the Th17 phenotype. In addition, the immune response of the perifollicular infiltrate depends on factors produced by the sebaceous glands, mostly sebaceous lipids. Human sebocytes in vitro express functional pattern recognition receptors, which are likely to interact with bacteria in acne pathogenesis. Sex steroids, peroxisome proliferator-activated receptor ligands, neuropeptides, endocannabinoids and a selective apoptotic process contribute to a complex regulation of sebocyte-induced immunological reaction in numerous acquired and congenital skin diseases, including hair diseases and atopic dermatitis.


Assuntos
Acne Vulgar , Dermatite Atópica , Adulto , Adolescente , Humanos , Imunidade Inata , Homeostase , Dermatite Atópica/complicações , Lipídeos
2.
Development ; 149(14)2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35815643

RESUMO

The barrier-forming, self-renewing mammalian epidermis comprises keratinocytes, pigment-producing melanocytes and resident immune cells as first-line host defense. In murine tail skin, interfollicular epidermis patterns into pigmented 'scale' and hypopigmented 'interscale' epidermis. Why and how mature melanocytes accumulate in scale epidermis is unresolved. Here, we delineate a cellular hierarchy among epidermal cell types that determines skin patterning. Already during postnatal development, melanocytes co-segregate with newly forming scale compartments. Intriguingly, this process coincides with partitioning of both Langerhans cells and dendritic epidermal T cells to interscale epidermis, suggesting functional segregation of pigmentation and immune surveillance. Analysis of non-pigmented mice and of mice lacking melanocytes or resident immune cells revealed that immunocyte patterning is melanocyte and melanin independent and, vice versa, immune cells do not control melanocyte localization. Instead, genetically enforced progressive scale fusion upon Lrig1 deletion showed that melanocytes and immune cells dynamically follow epithelial scale:interscale patterns. Importantly, disrupting Wnt-Lef1 function in keratinocytes caused melanocyte mislocalization to interscale epidermis, implicating canonical Wnt signaling in organizing the pigmentation pattern. Together, this work uncovers cellular and molecular principles underlying the compartmentalization of tissue functions in skin.


Assuntos
Epiderme , Cauda , Animais , Células Epidérmicas/metabolismo , Epiderme/metabolismo , Queratinócitos/metabolismo , Mamíferos/metabolismo , Melaninas/metabolismo , Melanócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Pigmentação da Pele , Cauda/metabolismo
3.
EMBO Rep ; 22(10): e52301, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34342114

RESUMO

Maintaining the architecture, size and composition of an intact stem cell (SC) compartment is crucial for tissue homeostasis and regeneration throughout life. In mammalian skin, elevated expression of the anti-apoptotic Bcl-2 protein has been reported in hair follicle (HF) bulge SCs (BSCs), but its impact on SC function is unknown. Here, we show that systemic exposure of mice to the Bcl-2 antagonist ABT-199/venetoclax leads to the selective loss of suprabasal BSCs (sbBSCs), thereby disrupting cyclic HF regeneration. RNAseq analysis shows that the pro-apoptotic BH3-only proteins BIM and Bmf are upregulated in sbBSCs, explaining their addiction to Bcl-2 and the marked susceptibility to Bcl-2 antagonism. In line with these observations, conditional knockout of Bcl-2 in mouse epidermis elevates apoptosis in BSCs. In contrast, ectopic Bcl-2 expression blocks apoptosis during HF regression, resulting in the accumulation of quiescent SCs and delaying HF growth in mice. Strikingly, Bcl-2-induced changes in size and composition of the HF bulge accelerate tumour formation. Our study identifies a niche-instructive mechanism of Bcl-2-regulated apoptosis response that is required for SC homeostasis and tissue regeneration, and may suppress carcinogenesis.


Assuntos
Proteínas Reguladoras de Apoptose , Folículo Piloso , Animais , Apoptose/genética , Camundongos , Pele , Células-Tronco
4.
J Invest Dermatol ; 141(11): 2602-2610.e3, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33965403

RESUMO

The skin epidermis is attached to the underlying dermis by a laminin 332 (Lm332)-rich basement membrane. Consequently, loss of Lm332 leads to the severe blistering disorder epidermolysis bullosa junctionalis in humans and animals. Owing to the indispensable role of Lm332 in keratinocyte adhesion in vivo, the severity of the disease has limited research into other functions of the protein. We have conditionally disrupted Lm332 expression in basal keratinocytes of adult mice. Although blisters develop along the interfollicular epidermis, hair follicle basal cells provide sufficient anchorage of the epidermis to the dermis, making inducible deletion of the Lama3 gene compatible with life. Loss of Lm332 promoted the thickening of the epidermis and exaggerated desquamation. Global RNA expression analysis revealed major changes in the expression of keratins, cornified envelope proteins, and cellular stress markers. These modifications of the keratinocyte genetic program are accompanied by changes in cell shape and disorganization of the actin cytoskeleton. These data indicate that loss of Lm332-mediated progenitor cell adhesion alters cell fate and disturbs epidermal homeostasis.


Assuntos
Moléculas de Adesão Celular/fisiologia , Homeostase/fisiologia , Queratinócitos/citologia , Citoesqueleto de Actina/fisiologia , Alarminas/fisiologia , Animais , Vesícula/etiologia , Diferenciação Celular , Epiderme/patologia , Queratinas/análise , Camundongos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/análise , Calinina
5.
Exp Dermatol ; 30(4): 588-597, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33599012

RESUMO

Sebaceous glands (SGs), typically associated with hair follicles, are critical for the homeostasis and function of mammalian skin. The main physiological function of SGs is the production and holocrine secretion of sebum to lubricate and protect the skin. Defective SGs have been linked to a variety of skin disorders, including acne, seborrheic dermatitis and formation of sebaceous tumors. Thus, a better understanding how SGs are formed and maintained is important to unravel the underlying molecular and cellular mechanisms of SG pathologies and to find better and effective therapies. Over the last two decades, research has come a long way from the initial identification of skin epithelial stem cells to the isolation and functional characterization of multiple stem cell pools as well as a better understanding of their unique and complex activities that drive skin homeostasis and operate in skin pathologies. Here, we discuss recent progress in unravelling cellular mechanisms underlying SG development, homeostasis and sebaceous tumor formation and assess the role of stem and progenitor cells in controlling SG physiology and disease processes. The development of elegant in vivo imaging as well as various in vitro and ex vivo stem cell and SG tissue models will advance mechanistic studies on SG function and allow drug screening and testing for efficient and successful targeting SG pathologies.


Assuntos
Folículo Piloso/patologia , Homeostase , Glândulas Sebáceas/patologia , Dermatopatias/patologia , Células-Tronco/patologia , Animais , Humanos , Camundongos
6.
Nat Commun ; 9(1): 3425, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143626

RESUMO

Transcription factors ensure skin homeostasis via tight regulation of distinct resident stem cells. Here we report that JunB, a member of the AP-1 transcription factor family, regulates epidermal stem cells and sebaceous glands through balancing proliferation and differentiation of progenitors and by suppressing lineage infidelity. JunB deficiency in basal progenitors results in a dermatitis-like syndrome resembling seborrheic dermatitis harboring structurally and functionally impaired sebaceous glands with a globally altered lipid profile. A fate switch occurs in a subset of JunB deficient epidermal progenitors during wound healing resulting in de novo formation of sebaceous glands. Dysregulated Notch signaling is identified to be causal for this phenotype. In fact, pharmacological inhibition of Notch signaling can efficiently restore the lineage drift, impaired epidermal differentiation and disrupted barrier function in JunB conditional knockout mice. These findings define an unprecedented role for JunB in epidermal-pilosebaceous stem cell homeostasis and its pathology.


Assuntos
Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Epiderme/metabolismo , Camundongos , Camundongos Knockout , Glândulas Sebáceas/citologia , Glândulas Sebáceas/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Cicatrização/genética , Cicatrização/fisiologia
7.
EMBO J ; 36(2): 151-164, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-27940653

RESUMO

Understanding how complex tissues are formed, maintained, and regenerated through local growth, differentiation, and remodeling requires knowledge on how single-cell behaviors are coordinated on the population level. The self-renewing hair follicle, maintained by a distinct stem cell population, represents an excellent paradigm to address this question. A major obstacle in mechanistic understanding of hair follicle stem cell (HFSC) regulation has been the lack of a culture system that recapitulates HFSC behavior while allowing their precise monitoring and manipulation. Here, we establish an in vitro culture system based on a 3D extracellular matrix environment and defined soluble factors, which for the first time allows expansion and long-term maintenance of murine multipotent HFSCs in the absence of heterologous cell types. Strikingly, this scheme promotes de novo generation of HFSCs from non-HFSCs and vice versa in a dynamic self-organizing process. This bidirectional interconversion of HFSCs and their progeny drives the system into a population equilibrium state. Our study uncovers regulatory dynamics by which phenotypic plasticity of cells drives population-level homeostasis within a niche, and provides a discovery tool for studies on adult stem cell fate.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Folículo Piloso/citologia , Técnicas de Cultura de Órgãos/métodos , Células-Tronco/fisiologia , Animais , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
J Invest Dermatol ; 136(12): 2406-2416, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27452221

RESUMO

Partitioning-defective (Par) proteins contribute to multiprotein complexes that drive cell polarity and fate in invertebrates. Of these, the ternary Par3-atypical protein kinase C-Par6 polarity complex mediates asymmetry in various systems, whereas Par3 and aPKC/Par6 can also act independently. aPKC-λ has recently been implicated in epidermal differentiation and stem cell fate; however, whether Par3 contributes to the homeostasis of adult stratified epithelia is currently unknown. Here, we provide functional evidence that epidermal Par3 loss disturbed the inside-out skin barrier, coinciding with altered expression and localization of principle tight junction components, and that epidermal differentiation and thickness were increased. Moreover, Par3 inactivation caused an initial expansion and later decline of hair follicle bulge stem cells, accompanied by an enrichment of committed progenitors, formation of hypertrophic sebaceous glands, and increased epidermal differentiation, suggesting aberrant cell fate decisions. Importantly, and opposite to aPKCλ deletion, Par3 loss did not enhance perpendicular cell divisions. Instead, in Par3-deficient hair follicles, spindles were shifted toward planar orientation, indicating that abnormal differentiation after Par3 inactivation is unlikely to be attributed to increased perpendicular spindle orientation. Collectively, mammalian Par3 controls the epidermal barrier, differentiation, and stem cell maintenance in the pilosebaceous unit, which are all essential for the homeostasis of an important barrier-forming epithelium.


Assuntos
Polaridade Celular/fisiologia , Homeostase/fisiologia , Queratinócitos/citologia , Proteína Quinase C/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Epiderme/metabolismo , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Papel (figurativo) , Células-Tronco/metabolismo
9.
Nat Commun ; 6: 5874, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25608467

RESUMO

Epithelial cancer constitutes a major clinical challenge and molecular mechanisms underlying the process of tumour initiation are not well understood. Here we demonstrate that hair follicle bulge stem cells (SCs) give rise to well-differentiated sebaceous tumours and show that SCs are not only crucial in tumour initiation, but are also involved in tumour plasticity and heterogeneity. Our findings reveal that SC-specific expression of mutant Lef1, which mimics mutations found in human sebaceous tumours, drives sebaceous tumour formation. Mechanistically, we demonstrate that mutant Lef1 abolishes p53 activity in SCs. Intriguingly, mutant Lef1 induces DNA damage and interferes with SC-specific gatekeeper functions normally protecting against accumulations of DNA lesions and cell loss. Thus, normal control of SC proliferation is disrupted by mutant Lef1, thereby allowing uncontrolled propagation of tumour-initiating SCs. Collectively, these findings identify underlying molecular and cellular mechanisms of tumour-initiating events in tissue SCs providing a potential target for future therapeutic strategies.


Assuntos
Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neoplasias das Glândulas Sebáceas/patologia , Neoplasias Cutâneas/patologia , Células-Tronco/citologia , Proteína Supressora de Tumor p53/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animais , Animais Recém-Nascidos , Apoptose , Carcinogênese , Carcinógenos , Linhagem da Célula , Proliferação de Células , Separação Celular , Transformação Celular Neoplásica/patologia , Cruzamentos Genéticos , Dano ao DNA , Progressão da Doença , Epiderme/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Neoplasias das Glândulas Sebáceas/metabolismo , Neoplasias Cutâneas/metabolismo , Raios Ultravioleta
10.
J Invest Dermatol ; 135(3): 679-689, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25371971

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Folículo Piloso/crescimento & desenvolvimento , Mitocôndrias/fisiologia , Morfogênese/fisiologia , Fenômenos Fisiológicos da Pele , Animais , Apoptose/fisiologia , Proliferação de Células , DNA Mitocondrial/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Metabolismo Energético/fisiologia , Epitélio/fisiologia , Folículo Piloso/citologia , Proteínas de Grupo de Alta Mobilidade/deficiência , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/fisiologia , Camundongos , Camundongos Knockout , Modelos Animais
11.
Exp Dermatol ; 23(12): 881-3, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25039641

RESUMO

Wnt/ß-catenin signalling is a key regulator of hair follicle (HF) morphogenesis and life-long HF regeneration. In a recently published issue of Experimental Dermatology, Lei et al. report that sustained WNT10B supply and pathway activation in regenerating mouse HF increased the width of hair bulbs, hair shafts and the dermal papilla (DP), and enlarged the CD34(+) HF bulge cell compartment. Notably, WNT10B affected primarily zigzag HFs, while size and morphology of other HF types remained largely unaffected. Thus, these findings raise a number of questions regarding a HF type-specific function of Wnt/ß-catenin and on the role of the WNT-stimulated DP in this process.


Assuntos
Folículo Piloso/metabolismo , Via de Sinalização Wnt , Animais , Antígenos CD34/metabolismo , Folículo Piloso/anatomia & histologia , Folículo Piloso/crescimento & desenvolvimento , Camundongos , Morfogênese , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
12.
Mol Cell Biol ; 34(16): 3086-95, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24891618

RESUMO

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.


Assuntos
Fator de Crescimento Epidérmico/biossíntese , Receptores ErbB/biossíntese , Glândulas Sebáceas/embriologia , Glândulas Sebáceas/patologia , Animais , Fator de Crescimento Epidérmico/genética , Epiderme/crescimento & desenvolvimento , Epiderme/patologia , Epigen , Receptores ErbB/genética , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/patologia , Hiperplasia/metabolismo , Queratinócitos/metabolismo , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Glândulas Sebáceas/metabolismo , Sebo/metabolismo , Transdução de Sinais/genética
13.
Cell Rep ; 4(2): 340-51, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23871669

RESUMO

Mutations in the hedgehog pathway drive the formation of tumors in many different organs, including the development of basal cell carcinoma in the skin. However, little is known about the role of epidermal Indian hedgehog (Ihh) in skin physiology. Using mouse genetics, we identified overlapping and distinct functions of Ihh in different models of epidermal tumorigenesis. Epidermal deletion of Ihh resulted in increased formation of benign squamous papilloma. Strikingly, Ihh-deficient mice showed an increase in malignant squamous cell carcinoma and developed lung and lymph node metastases. In a sebaceous gland tumor model, Ihh deficiency inhibited tumor cell differentiation. More mechanistically, IHH stimulated cell proliferation by activating the transcription factor GLI2 in human keratinocytes and human tumors. Thus, our results uncover important functions for Ihh signaling in controlling proliferation, differentiation, malignant progression, and metastasis of epithelial cancer, establishing Ihh as a gatekeeper for controlling the grade of tumor malignancy.


Assuntos
Transformação Celular Neoplásica/patologia , Proteínas Hedgehog/metabolismo , Neoplasias Cutâneas/patologia , Pele/patologia , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Proteínas Hedgehog/genética , Humanos , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Pele/metabolismo , Neoplasias Cutâneas/metabolismo , Transfecção
14.
Methods Mol Biol ; 989: 45-60, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23483386

RESUMO

Lineage tracing of tissue stem cells represents a powerful tool to address fundamental questions of deve-lopment, differentiation and cellular renewal in a natural tissue environment. The Cre/lox site-specific recombination system is increasingly used to genetically label specific cell populations to perform cell lineage tracing or fate mapping experiments in sophisticated mouse models. Here we describe a method of labeling and subsequent tracking stem cells of the hair follicle bulge region in mouse skin. Hair follicle stem cells are specifically labeled by expressing the Cre recombinase under control of keratin15 (K15) regulatory sequences and by crossing the Cre-containing animals with Cre-sensitive Rosa26R (R26R) reporter mice. To achieve a temporal control of recombinase activity in stem cells, Cre is fused to a modified estrogen receptor (CreER(G)T2). In the K15CreER(G)T2/R26R mouse model, hair follicle stem cells (HFSCs) are specifically labeled after Cre activation upon treatment of mice with tamoxifen. By analyzing the skin tissue at different time points following genetic labeling, important information on stem cell behavior and contribution of labeled stem cells to epidermal structures during tissue homeostasis and hair follicle regeneration are obtained. Combining the lineage tracing approach with the whole mount technique allows examining large areas of the epidermis containing many hair follicles and sebaceous glands and reveals the complex three-dimensional relationship of labeled stem cell clones within the tissue.


Assuntos
Células Epidérmicas , Folículo Piloso/citologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Células Cultivadas , Epiderme/efeitos dos fármacos , Folículo Piloso/efeitos dos fármacos , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pele/citologia , Pele/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Tamoxifeno
15.
Semin Cell Dev Biol ; 23(8): 928-36, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22960253

RESUMO

The important role of epidermal appendages especially the sebaceous gland has only recently been recognized. In particular, it has been convincingly shown that normal development and maintenance of the sebaceous gland are required for skin homeostasis since atrophic sebaceous glands and disturbances in sebaceous lipid composition result in major defects of the physiological barrier and maintenance of the skin. Consequently, it is important to unravel the signaling network controlling proper sebaceous lineage differentiation in mammalian skin and to understand the underlying mechanisms leading to severe skin diseases, including abnormal proliferation and differentiation of the gland, defects of the lipid metabolism and barrier, as well as sebaceous tumor formation. Over the last years, results from transgenic and knock out mouse models manipulating distinct signaling pathways in the skin as well as the detailed analysis of human sebaceous gland-derived cell lines provided new insights into crucial mediators balancing proliferation and differentiation of the sebaceous gland. Here, we discuss our current knowledge of in vivo mechanisms of sebaceous gland development, maintenance and disorders and highlight recent contributions to the field of sebaceous gland biology.


Assuntos
Homeostase , Glândulas Sebáceas/metabolismo , Animais , Humanos , Glândulas Sebáceas/crescimento & desenvolvimento , Transdução de Sinais , Dermatopatias/metabolismo , Dermatopatias/patologia , Transcrição Gênica
16.
J Cell Sci ; 125(Pt 4): 896-905, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22275433

RESUMO

The small GTPase Rac1 is ubiquitously expressed in proliferating and differentiating layers of the epidermis and hair follicles. Previously, Rac1 was shown to regulate stem cell behaviour in these compartments. We have asked whether Rac1 has, in addition, a specific, stem-cell-independent function in the regulation of terminal hair follicle differentiation. To address this, we have expressed a constitutively active mutant of Rac1, L61Rac1, only in the basal epidermal layer and outer root sheath of mice possessing an epidermis-specific deletion of endogenous Rac1, which experience severe hair loss. The resulting 'rescue' mice exhibited a hair coat throughout their lives. Therefore, expression of Rac1 activity in the keratin-14-positive compartment of the skin is sufficient for the formation of hair follicles and hair in normal quantities. The quality of hair formed in rescue mice was, however, not normal. Rescue mice showed a grey, dull hair coat, whereas that of wild-type and L61Rac1-transgenic mice was black and shiny. Hair analysis in rescue mice revealed altered structures of the hair shaft and the cuticle and disturbed organization of medulla cells and pigment distribution. Disorganization of medulla cells correlates with the absence of cortical, keratin-filled spikes that normally protrude from the cortex into the medulla. The desmosomal cadherin Dsc2, which normally decorates these protrusions, was found to be reduced or absent in the hair of rescue mice. Our study demonstrates regulatory functions for Rac1 in the formation of hair structure and pigmentation and thereby identifies, for the first time, a role for Rac1 in terminal differentiation.


Assuntos
Diferenciação Celular , Cor de Cabelo/fisiologia , Cabelo/citologia , Cabelo/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Desmocolinas , Feminino , Cabelo/crescimento & desenvolvimento , Cabelo/patologia , Folículo Piloso/crescimento & desenvolvimento , Humanos , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , Fatores de Tempo , Transgenes
17.
Dev Biol ; 363(1): 138-46, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22227295

RESUMO

The hair follicle (HF) and the sebaceous gland (SG) constitute the two integral parts of the pilosebaceous unit and significantly contribute to the barrier function of mammalian skin. Considerable progress has been made in our understanding how HF formation is regulated. However, the development of the SG is poorly understood, both at the molecular and cellular level. Here, we investigate the process of SG morphogenesis and the dynamics of its cellular organisation in more detail. The spatial and temporal organisation of distinct stem and progenitor compartments was analysed during morphogenesis of the pilosebaceous unit in mouse tail epidermis. Our experiments reveal a dynamic expression pattern for diverse HF stem cell marker molecules including Sox9 and Lrig1. Surprisingly, Sox9 and Lrig1 are initially coexpressed by epidermal progenitor cells and are confined to different regions within the pilosebaceous unit when the specification of the sebocyte cell lineage takes place. We demonstrate that SG development at the distal part of the HF is driven by asymmetric cell fate decision of Lrig1 positive stem cells, whereas MTS24/Plet1 positive precursor cells seem not to play a role in this process. Importantly, our data clearly show that distinct stem and progenitor compartments are established at different time points of development. By studying the process of SG morphogenesis more precisely, we discovered that the two prominent SGs attached to one tail HF originate from one small cluster of sebocyte cells. Finally, we show regional specificity for HF patterning and spatio-temporal control of the underlying molecular signals initiating the development of the pilosebaceous unit.


Assuntos
Morfogênese , Glândulas Sebáceas/metabolismo , Pele/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Epiderme/embriologia , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Imunofluorescência , Folículo Piloso/embriologia , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Cinética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas da Gravidez/metabolismo , Fatores de Transcrição SOX9/metabolismo , Glândulas Sebáceas/embriologia , Glândulas Sebáceas/crescimento & desenvolvimento , Pele/embriologia , Pele/crescimento & desenvolvimento , Cauda , Fatores de Tempo
18.
J Invest Dermatol ; 132(2): 337-45, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21938009

RESUMO

Defects in the function of the skin barrier are associated with a wide variety of skin diseases, many of which are not well characterized at the molecular level. Using Lef1 (lymphoid enhancer-binding factor 1) dominant-negative mutant mice, we demonstrate here that altered epidermal TCF (T cell factor)/Lef1 signaling results in severe impairment of the stratum corneum skin barrier and early postnatal death. Barrier defects were accompanied by major changes in lipid composition and ultrastructural abnormalities in assembly and extrusion of lipid lamellae of the interfollicular epidermis, as well as abnormal processing of profilaggrin. In contrast, tight-junction formation and stratified organization of the interfollicular epidermis was not obviously disturbed in Lef1 mutant mice. Molecular analysis revealed that TCF/Lef1 signaling regulates expression of lipid-modifying enzymes, such as Elovl3 and stearoyl coenzyme A desaturase 1 (SCD1), which are key regulators of barrier function. Promoter analysis and chromatin immunoprecipitation experiments indeed showed that SCD1 is a direct target of Lef1. Together, our data demonstrate that functional TCF/Lef1 signaling governs important aspects of epidermal differentiation and lipid metabolism, thereby regulating skin barrier function.


Assuntos
Metabolismo dos Lipídeos , Fator 1 de Ligação ao Facilitador Linfoide/fisiologia , Pele/metabolismo , Animais , Lipídeos/análise , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Pele/citologia , Fatores de Transcrição TCF , Junções Íntimas/fisiologia
19.
EMBO J ; 30(15): 3004-18, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21694721

RESUMO

Mammalian epidermis consists of the interfollicular epidermis, hair follicles (HFs) and associated sebaceous glands (SGs). It is constantly renewed by stem and progenitor cell populations that have been identified and each compartment features a distinct mechanism of cellular turnover during renewal. The functional relationship between the diverse stem cell (SC) pools is not known and molecular signals regulating the establishment and maintenance of SC compartments are not well understood. Here, we performed lineage tracing experiments to demonstrate that progeny of HF bulge SCs transit through other SC compartments, suggesting a hierarchy of competent multipotent keratinocytes contributing to tissue renewal. The bulge was identified as a bipotent SC compartment that drives both cyclic regeneration of HFs and continuous renewal of SGs. Our data demonstrate that aberrant signalling by TCF/Lef1, transcription factors crucial for bulge SC activation and hair differentiation, results in development of ectopic SGs originating from bulge cells. This process of de novo SG formation is accompanied by the establishment of new progenitor niches. Detailed molecular analysis suggests the recapitulation of steps of tissue morphogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Folículo Piloso/citologia , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Células-Tronco/fisiologia , Animais , Expressão Gênica , Glicoproteínas de Membrana/biossíntese , Camundongos , Modelos Biológicos , Proteínas do Tecido Nervoso/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Glândulas Sebáceas/citologia
20.
Am J Pathol ; 174(6): 2116-28, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19389925

RESUMO

Wound healing is a crucial regenerative process in all organisms. We examined expression, integrity, and function of the proteins in the hepatocyte growth factor (HGF)/c-Met signaling pathway in normally healing and non-healing human skin wounds. Whereas in normally healing wounds phosphorylation of c-Met was most prominent in keratinocytes and dermal cells, in non-healing wounds phosphorylation of c-Met was barely detectable, suggesting reduced c-Met activation. In wound exudates obtained from non-healing, but not from healing wounds, HGF protein was a target of substantial proteolytic processing that was different from the classical activation by known serine proteases. Western blot analysis and protease inhibitor studies revealed that HGF is a target of neutrophil elastase and plasma kallikrein during skin repair. Proteolytic processing of HGF by each of these proteases significantly attenuated keratinocyte proliferation, wound closure capacity in vitro, and c-Met signal transduction. Our findings reveal a novel pathway of HGF processing during skin repair. Conditions in which proteases are imbalanced and tend toward increased proteolytic activity, as in chronic non-healing wounds, might therefore compromise HGF activity due to the inactivation of the HGF protein and/or the generation of HGF fragments that ultimately mediate a dominant negative effect and limit c-Met activation.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Transdução de Sinais/fisiologia , Cicatrização/fisiologia , Idoso , Western Blotting , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Exsudatos e Transudatos/química , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-met/metabolismo , Serina Endopeptidases/metabolismo , Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...