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










Publication year range
1.
Br J Dermatol ; 148(1): 30-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12534591

ABSTRACT

BACKGROUND: Adrenomedullin (AM) is a regulatory peptide that is synthesized and secreted by a wide number of cells and tissues. AM is a potent vasodilator, but also exerts other functions, such as regulating cell growth and antimicrobial defence. Two receptors, L1 and calcitonin receptor-like receptor (CRLR), which are able to bind AM, have been cloned and characterized. OBJECTIVES: To investigate expression of AM protein and its receptors in human skin and during different stages of the human hair cycle and, moreover, because of the suggested antimicrobial function of AM in skin, to investigate AM immunoreactivity (IR) in inflammatory acne lesions compared with healthy pilosebaceous follicles. METHODS: We used immunohistochemistry to determine the distribution of AM and its receptors in human skin and during different stages of the human hair cycle. AM IR in inflammatory acne lesions was investigated to evaluate the antimicrobial function of the protein, and hair follicle cultures were performed to examine the role of AM in differentiation and proliferation of hair follicle keratinocytes. RESULTS: Strong IR for AM and its receptors was present in the suprabasal epidermis, in the melanocytes of the epidermis, and in sweat and sebaceous glands. In the hair follicle, AM protein was strongly expressed in the basal and suprabasal layers of the hair bulb and the proximal outer root sheath (ORS). In the distal ORS, AM expression was increasingly suprabasal, especially in proximity to the bulge region where the basal cell layer was free of IR. IR for the CRLR revealed a similar expression pattern to that seen for AM. In contrast, L1 IR showed a suprabasal pattern of IR throughout the ORS. Similar expression of AM and its receptors was observed in catagen and early anagen follicles. AM expression was not markedly upregulated in acne lesions, suggesting a minor role for this antimicrobial peptide in acne. Despite its well-documented mitogenic effects, particularly in oral and skin keratinocytes, AM had no significant effect on hair follicle growth in vitro. CONCLUSIONS: AM and its receptors are expressed in human hair follicles, and both AM and its receptors are colocalized in the same compartments and cell types of the skin. This finding is consistent with the proposed autocrine/paracrine mechanism in the physiology of AM.


Subject(s)
Acne Vulgaris/metabolism , Peptides/metabolism , Skin/metabolism , Adrenomedullin , Blotting, Western , Calcitonin Receptor-Like Protein , Cells, Cultured , Hair Follicle/drug effects , Hair Follicle/growth & development , Hair Follicle/metabolism , Humans , Immunoenzyme Techniques , Peptides/pharmacology , Peptides/physiology , Receptors, Adrenomedullin , Receptors, Calcitonin/metabolism , Receptors, Peptide/metabolism , Scalp/metabolism , Skin/growth & development , Up-Regulation
2.
J Invest Dermatol ; 117(5): 1120-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11710922

ABSTRACT

A rich residential microflora is harboured by the distal outer root sheath of the hair follicle and the hair canal - normally without causing skin diseases. Although the basic mechanisms involved in the development of inflammation during acne vulgaris remain unclear, microbial agents might play an important role in this process. In this study we have analyzed by in situ hybridization and immunohistochemistry the expression patterns of two antimicrobial peptides, human beta defensin-1 and human beta defensin-2, in healthy human hair follicles as well as in perilesional and intralesional skin of acne vulgaris lesions such as comedones, papules, and pustules. Strong defensin-1 and defensin-2 immunoreactivity was found in all suprabasal layers of the epidermis, the distal outer root sheath of the hair follicle, and the pilosebaceous duct. Marked defensin-1 and defensin-2 immunoreactivity was also found in the sebaceous gland and in the basal layer of the central outer root sheath including the bulge region. The majority of acne biopsies displayed a marked upregulation of defensin-2 immunoreactivity in the lesional and perilesional epithelium - in particular in pustules - and a less marked upregulation of defensin-1 immunoreactivity. The upregulation of beta-defensin expression in acne vulgaris lesions compared to controls suggests that beta-defensins may be involved in the pathogenesis of acne vulgaris.


Subject(s)
Acne Vulgaris/metabolism , Hair Follicle/metabolism , Skin/metabolism , beta-Defensins/metabolism , Acne Vulgaris/pathology , Humans , Immunohistochemistry , In Situ Hybridization , RNA, Messenger/metabolism , Reference Values , Scalp/metabolism , Tissue Distribution , Up-Regulation , beta-Defensins/genetics
3.
J Invest Dermatol ; 117(1): 3-15, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11442744

ABSTRACT

Numerous strains of mice with defined mutations display pronounced abnormalities of hair follicle cycling, even in the absence of overt alterations of the skin and hair phenotype; however, in order to recognize even subtle, hair cycle-related abnormalities, it is critically important to be able to determine accurately and classify the major stages of the normal murine hair cycle. In this comprehensive guide, we present pragmatic basic and auxiliary criteria for recognizing key stages of hair follicle growth (anagen), regression (catagen) and quiescence (telogen) in C57BL/6NCrlBR mice, which are largely based on previous work from other authors. For each stage, a schematic drawing and representative micrographs are provided in order to illustrate these criteria. The basic criteria can be employed for all mouse strains and require only routine histochemical techniques. The auxiliary criteria depend on the immunohistochemical analysis of three markers (interleukin-1 receptor type I, transforming growth factor-beta receptor type II, and neural cell-adhesion molecule), which allow a refined analysis of anatomical hair follicle compartments during all hair cycle stages. In contrast to prior staging systems, we suggest dividing anagen III into three distinct substages, based on morphologic differences, onset and progression of melanogenesis, and the position of the dermal papilla in the subcutis. The computer-generated schematic representations of each stage are presented with the aim of standardizing reports on follicular gene and protein expression patterns. This guide should become a useful tool when screening new mouse mutants or mice treated with pharmaceuticals for discrete morphologic abnormalities of hair follicle cycling in a highly reproducible, easily applicable, and quantifiable manner.


Subject(s)
Dermatology/standards , Hair Follicle/anatomy & histology , Hair Follicle/growth & development , Animals , Guidelines as Topic , Mice
4.
J Invest Dermatol ; 116(6): 947-55, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11407986

ABSTRACT

In this study, we have correlated cutaneous apoptosis and proliferation in neonatal mice during hair follicle morphogenesis. We have applied a novel triple- staining technique that uses Ki67 immunoreactivity as a marker of proliferation as well as TUNEL and Hoechst 33342 staining as apoptosis markers. We have also assessed the immunoreactivity of interleukin-1 beta-converting enzyme, caspase 1, a key enzyme in the execution of apoptosis, and of P-cadherin, which has been suggested as a key adhesion receptor in segregating proliferating keratinocytes. The TUNEL data were systematically compared with high resolution light microscopy and transmission electron microscopy data. Virtually all keratinocytes of the developing hair bud were strongly Ki67(+), suggesting that the hair bud is not an epidermal invagination but primarily the product of localized keratinocyte proliferation. As hair follicle development advanced, three distinct foci of proliferation became apparent: the distal outer root sheath around the hair canal, the mid outer root sheath, and the proximal hair matrix. Of these proliferating hair follicle keratinocytes only defined subsets expressed P-cadherin. TUNEL(+) cells in the hair follicle were not found before stage 5 of murine hair follicle morphogenesis. During the early stages of hair follicle development, interleukin-1 beta-converting enzyme immunoreactivity was present on all keratinocytes, but virtually disappeared from the proximal hair follicle epithelium later on. High resolution light microscopy/transmission electron microscopy revealed scattered and clustered apoptotic keratinocytes in all epithelial hair follicle compartments throughout hair follicle development, including its earliest stages. This highlights striking differences in the demarcation of apoptotic hair follicle keratinocytes between the TUNEL technique and high resolution light microscopy/transmission electron microscopy and suggests a role for apoptosis in sculpting the hair follicle even during early hair follicle development.


Subject(s)
Apoptosis , Hair Follicle/growth & development , Animals , Cadherins/analysis , Caspase 1/analysis , Cell Division , Female , Hair Follicle/chemistry , Hair Follicle/ultrastructure , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Morphogenesis
5.
J Invest Dermatol ; 116(4): 617-22, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11286633

ABSTRACT

The mammalian hair follicle is a highly dynamic skin appendage that undergoes repeated cycles of growth and regression, involving closely co-ordinated regulation of cell proliferation, differentiation, and apoptosis. The Myc superfamily of transcription factors have been strongly implicated in the regulation of these processes in many tissues. Using immunohistochemistry, we have investigated the patterns of c-Myc, N-Myc, Max, and Mad1-4 expression at different stages of the human hair growth cycle. N-Myc, Max, Mad1, and Mad3 immunoreactivity was detected in the epidermis and the epithelium of both anagen and telogen hair follicles. Three distinct patterns of hair follicle c-Myc immunoreactivity were observed. In the infundibulum, c-Myc staining was predominantly in the basal layers, with little detectable immunoreactivity in the terminally differentiating suprabasal layers; this pattern was similar to that seen in the epidermis. In contrast, c-Myc expression in the follicle bulb was found both in the proliferating germinative epithelial cells and in the terminally differentiating matrix cells that give rise to the hair fiber. Finally, intense c-Myc immunoreactivity was detected in the bulge region of the outer root sheath. Using the C8/144B antibody as a bulge marker, we confirmed that c-Myc immunoreactivity in the outer root sheath correlates with the putative hair follicle stem cell compartment. c-Myc expression in the bulge was independent of the hair growth cycle stage. Our data suggest that Myc superfamily members serve different functions in separate epithelial compartments of the hair follicle and may play an important role in determining cell fate within the putative stem cell compartment.


Subject(s)
Genes, myc , Hair Follicle/metabolism , Hair/growth & development , Multigene Family , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Cell Cycle Proteins , Cell Division , DNA-Binding Proteins/metabolism , Down-Regulation , Epithelium/metabolism , Hair Follicle/cytology , Humans , Immunohistochemistry , Nuclear Proteins , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Tissue Distribution
6.
Br J Dermatol ; 142(5): 862-73, 2000 May.
Article in English | MEDLINE | ID: mdl-10809841

ABSTRACT

The immunology of the hair follicle, its relationship with the 'skin immune system' and its role in hair diseases remain biologically intriguing and clinically important. In this study, we analysed the immunoreactivity patterns of 15 immunodermatological markers to determine the cellular composition and immune privilege of the human hair follicle immune system in anagen VI (growth phase). The most prominent cells located in or around the hair follicle were Langerhans cells, CD4+ or CD8+ T cells, macrophages and mast cells, whereas B cells, natural killer cells and gammadelta T cells were found very rarely. Langerhans cells (CD1a+, major histocompatibility complex, MHC class II+), and T cells (CD4+ or CD8+) were predominantly distributed in the distal hair follicle epithelium, whereas macrophages (CD68+, MHC class II+) and mast cells (Giemsa+) were located in the perifollicular connective tissue sheath. Transmission electron microscopy confirmed low numbers of immune cells in the proximal hair follicle epithelium, and very few macrophages and Langerhans cells were seen in the dermal papilla. Melanophages were observed in the connective tissue sheath and dermal papilla. MHC class I (HLA-A, -B, -C) and beta2-microglobulin immunoreactivity was found on most skin cells, but was substantially reduced on isthmus keratinocytes and virtually absent in the proximal hair follicle epithelium. Apart from the absence of Fas ligand immunoreactivity, the sharply reduced numbers of T cells and Langerhans cells, and the virtual absence of MHC class I expression all suggest that the anagen proximal hair follicle constitutes an area of immune privilege within the hair follicle immune system, whose collapse may be crucial for the pathogenesis of alopecia areata.


Subject(s)
Hair Follicle/immunology , Immunity, Cellular/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD20/analysis , B-Lymphocytes/immunology , Biomarkers/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hair Follicle/cytology , Histocompatibility Antigens Class I/immunology , Humans , Intercellular Adhesion Molecule-1/analysis , Keratinocytes/chemistry , Killer Cells, Natural/immunology , Langerhans Cells/immunology , Macrophages/immunology , Major Histocompatibility Complex/immunology , Mast Cells/immunology , Middle Aged , Receptors, Antigen, T-Cell, gamma-delta/analysis , Sebaceous Glands/immunology , Skin/immunology , T-Lymphocytes/chemistry , beta 2-Microglobulin/biosynthesis , fas Receptor/biosynthesis
7.
J Invest Dermatol ; 114(5): 909-16, 2000 May.
Article in English | MEDLINE | ID: mdl-10771470

ABSTRACT

After the completion of skin development, angiogenesis, i.e., the growth of new capillaries from pre-existing blood vessels, is held to occur in the skin only under pathologic conditions. It has long been noted, however, that hair follicle cycling is associated with prominent changes in skin perfusion, that the epithelial hair bulbs of anagen follicles display angiogenic properties, and that the follicular dermal papilla can produce angiogenic factors. Despite these suggestive observations, no formal proof is as yet available for the concept that angiogenesis is a physiologic event that occurs all over the mature mammalian integument whenever hair follicles switch from resting (telogen) to active growth (anagen). This study uses quantitative histomorphometry and double-immunohistologic detection techniques for the demarcation of proliferating endothelial cells, to show that synchronized hair follicle cycling in adolescent C57BL/6 mice is associated with substantial angiogenesis, and that inhibiting angiogenesis in vivo by the intraperitoneal application of a fumagillin derivative retards experimentally induced anagen development in these mice. Thus, angiogenesis is a physiologic event in normal postnatal murine skin, apparently is dictated by the hair follicle, and appears to be required for normal anagen development. Anagen-associated angiogenesis offers an attractive model for identifying the physiologic controls of cutaneous angiogenesis, and an interesting system for screening the effects of potential antiangiogenic drugs in vivo.


Subject(s)
Hair/growth & development , Neovascularization, Physiologic , Skin/blood supply , Animals , Cell Nucleus/ultrastructure , Cyclohexanes , Endothelium, Vascular/ultrastructure , Female , Mice , Mice, Inbred C57BL , O-(Chloroacetylcarbamoyl)fumagillol , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Sesquiterpenes/pharmacology
8.
Am J Pathol ; 156(4): 1395-405, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751363

ABSTRACT

Hair follicle (HF) growth and regression is an exquisitely regulated process of cell proliferation followed by massive cell death and is accompanied by cyclical expression of the apoptosis regulatory gene pair, Bcl-2 and Bax. To further investigate the role of Bcl-2 expression in the control of hair growth and keratinocyte apoptosis, we have used transgenic mice that overexpress human Bcl-2 in basal epidermis and in the outer root sheath under the control of the human keratin-14 promoter (K14/Bcl-2). When irradiated with ultraviolet B (UVB) light, K14/Bcl-2 mice developed about 5-10-fold fewer sunburn cells (ie, apoptotic keratinocytes) in the basal layer of the epidermis, compared to wild-type mice, whereas cultures of primary keratinocytes from transgenic mice were completely resistant to UVB-induced histone formation, at doses that readily induced histone release from wild-type cells. K14/Bcl-2 mice show no alteration of neonatal hair follicle morphogenesis or of the onset of the first wave of HF regression (catagen). However, compared to wild-type controls, K14/Bcl-2 mice subsequently displayed a significant acceleration of spontaneous catagen progression. During chemotherapy-induced alopecia, follicular dystrophy was promoted in K14/Bcl-2 mice. Thus, although K14-driven overexpression of Bcl-2 protected murine epidermal keratinocytes from UVB-induced apoptosis, it surprisingly promoted catagen- and chemotherapy-associated keratinocyte apoptosis.


Subject(s)
Alopecia/chemically induced , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Hair Follicle/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Ultraviolet Rays , Alopecia/pathology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Cyclophosphamide/pharmacology , Epidermis/pathology , Gene Expression , Hair Follicle/growth & development , Hair Follicle/physiopathology , Humans , Keratin-14 , Keratinocytes/drug effects , Keratinocytes/physiology , Keratinocytes/radiation effects , Keratins/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic/genetics , Phenotype , Proto-Oncogene Proteins c-bcl-2/genetics , Reference Values , Skin/physiopathology , Sunburn/pathology , Transgenes/physiology
9.
J Histochem Cytochem ; 48(4): 557-68, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727297

ABSTRACT

Although the intercellular adhesion molecule-1 (ICAM-1) is recognized for its pivotal role in inflammation and immune responses, its role in developmental systems, such as the cyclic growth (anagen) and regression (catagen) of the hair follicle, remains to be explored. Here we demonstrate that ICAM-1 expression in murine skin is even more widespread and more developmentally regulated than was previously believed. In addition to endothelial cells, selected epidermal and follicular keratinocyte subpopulations, as well as interfollicular fibroblasts, express ICAM-1. Murine hair follicles express ICAM-1 only late during morphogenesis. Thereafter, morphologically identical follicles markedly differ in their ICAM-1 expression patterns, which become strikingly hair cycle-dependent in both intra- and extrafollicular skin compartments. Minimal ICAM-1 and leukocyte function-associated (LFA-1) protein and mRNA expression is observed during early anagen and maximal expression during late anagen and catagen. Keratinocytes of the distal outer root sheath, fibroblasts of the perifollicular connective tissue sheath, and perifollicular blood vessels exhibit maximal ICAM-1 immunoreactivity during catagen, which corresponds to changes of LFA-1 expression on perifollicular macrophages. Finally, ICAM-1-deficient mice display significant catagen acceleration compared to wild-type controls. Therefore, ICAM-1 upregulation is not limited to pathological situations but is also important for skin and hair follicle remodeling. Collectively, this suggests a new and apparently nonimmunological function for ICAM-1-related signaling in cutaneous biology.


Subject(s)
Hair Follicle/growth & development , Hair Follicle/metabolism , Intercellular Adhesion Molecule-1/metabolism , Animals , Animals, Newborn , Hair Follicle/ultrastructure , Immunohistochemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/growth & development , Skin/metabolism , Skin/ultrastructure
10.
Am J Pathol ; 156(3): 1041-53, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10702420

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF), neurturin (NTN), and their receptors, GDNF family receptor alpha-1 (GFRalpha-1) and GDNF family receptor alpha-2 (GFRalpha-2), are critically important for kidney and nervous system development. However, their role in skin biology, specifically in hair growth control, is as yet unknown. We have studied expression and function of GDNF, neurturin, GFRalpha-1, and GFRalpha-2 in murine skin during the cyclic transformation of the hair follicle (HF) from its resting state (telogen) to active growth (anagen) and then through regression (catagen) back to telogen. GDNF protein and GFRalpha-1 messenger RNA are prominently expressed in telogen skin, which lacks NTN and GFRalpha-2 transcripts. Early anagen development is accompanied by a significant decline in the skin content of GDNF protein and GFRalpha-1 transcripts. During the anagen-catagen transition, GDNF, GFRalpha-1, NTN, and GFRalpha-2 transcripts reach maximal levels. Compared with wild-type controls, GFRalpha-1 (+/-) and GFRalpha-2 (-/-) knockout mice show a significantly accelerated catagen development. Furthermore, GDNF or NTN administration significantly retards HF regression in organ-cultured mouse skin. This suggests important, previously unrecognized roles for GDNF/GFRalpha-1 and NTN/GFRalpha-2 signaling in skin biology, specifically in the control of apoptosis-driven HF involution, and raises the possibility that GFRalpha-1/GFRalpha-2 agonists/antagonists might become exploitable for the treatment of hair growth disorders that are related to abnormalities in catagen development.


Subject(s)
Drosophila Proteins , Hair Follicle/growth & development , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Cells, Cultured , DNA/analysis , DNA Primers/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Hair Follicle/drug effects , Hair Removal , Heterozygote , Homozygote , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Neurturin , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism
11.
J Invest Dermatol ; 113(4): 523-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504436

ABSTRACT

Numerous spontaneous and experimentally induced mouse mutations develop a hair phenotype, which is often associated with more or less discrete abnormalities in hair follicle development. In order to recognize these, it is critically important to be able to determine and to classify accurately the major stages of normal murine hair follicle morphogenesis. As an aid, we propose a pragmatic and comprehensive guide, modified after previous suggestions by Hardy, and provide a list of easily recognizable classification criteria, illustrated by representative micrographs. Basic and more advanced criteria are distinguished, the former being applicable to all mouse strains and requiring only simple histologic stains (hematoxylin and eosin, Giemsa, periodic acid Schiff, alkaline phosphatase activity), the latter serving as auxiliary criteria, which require a pigmented mouse strain (like C57BL/6J) or immunohistochemistry (interleukin-1 receptor type I, transforming growth factor-beta receptor type II). In addition, we present simplified, computer-generated schematic drawings for the standardized recording and reporting of gene and antigen expression patterns during hair follicle development. This classification aid serves as a basic introduction into the field of hair follicle morphogenesis, aims at standardizing the presentation of related hair research data, and should become a useful tool when screening new mouse mutants for discrete abnormalities of hair follicle morphogenesis (compared with the respective wild type) in a highly reproducible, easily applicable, and quantifiable manner.


Subject(s)
Hair Follicle/embryology , Animals , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis , Phenotype , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interleukin-1/analysis , Receptors, Transforming Growth Factor beta/analysis
12.
Exp Dermatol ; 8(4): 237-46, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10439220

ABSTRACT

The role of adhesion molecules in the control of hair follicle (HF) morphogenesis, regression and cycling is still rather enigmatic. Since the adhesion molecules E- and P-cadherin (Ecad and Pcad) are functionally important, e.g. during embryonic pattern formation, we have studied their expression patterns during neonatal HF morphogenesis and cycling in C57/BL6 mice by immunohistology and semi-quantitative RT-PCR. The expression of both cadherins was strikingly hair cycle-dependent and restricted to distinct anatomical HF compartments. During HF morphogenesis, hair bud keratinocytes displayed strong Ecad and Pcad immunoreactivity (IR). While neonatal epidermis showed Ecad IR in all epidermal layers, Pcad IR was restricted to the basal layer. During later stages of HF morphogenesis and during anagen IV-VI of the adolescent murine hair cycle, the outer root sheath showed strong E- and Pcad IR. Instead, the outermost portion of the hair matrix and the inner root sheath displayed isolated Ecad IR, while the innermost portion of the hair matrix exhibited isolated Pcad IR. During telogen, all epidermal and follicular keratinocytes showed strong Ecad IR. This is in contrast to Pcad, whose IR was stringently restricted to matrix and secondary hair germ keratinocytes which are in closest proximity to the dermal papilla. These findings suggest that isolated or combined E- and/or Pcad expression is involved in follicular pattern formation by segregating HF keratinocytes into functionally distinct subpopulations; most notably, isolated Pcad expression may segregate those hair matrix keratinocytes into one functional epithelial tissue unit, which is particularly susceptible to growth control by dermal papilla-derived morphogens. The next challenge is to define which secreted agents implicated in hair growth control modulate these follicular cadherin expression patterns, and to define how these basic parameters of HF topobiology are altered during common hair growth disorders.


Subject(s)
Cadherins/metabolism , Hair Follicle/growth & development , Hair Follicle/metabolism , Aging/metabolism , Aging/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
13.
J Investig Dermatol Symp Proc ; 4(3): 226-34, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10674372

ABSTRACT

This paper delineates briefly why the immunology of the hair follicle matters (e.g., anti-infection defense, hair growth control by immunomodulatory agents, sequestration of follicular autoantigens), and which open key questions await clarification. We then focus on the murine hair follicle immune system (HIS) and its immune privilege. We show how the murine HIS is gradually constructed during hair follicle morphogenesis, and how it is transformed during hair follicle cycling. Key characteristics of the HIS are summarized, such as the absence of MHC class I expression in the anagen hair bulb and the very restricted distribution of antigen-presenting cells and intraepithelial T cells to the distal outer root sheath, which also expresses nonclassical MHC class Ib molecules. The interconnections between the HIS and the skin immune system (SIS) and potential hair growth-modulatory roles of mast cells and macrophages are addressed, and very recent findings on the human HIS are summarized. The paper closes by sketching immunobiologic, clinical, and pharmacologic perspectives in trichoimmunology that deserve the attention of immunologists, dermatologists, and hair biologists alike.


Subject(s)
Autoantigens/immunology , Hair Follicle/immunology , Animals , Hair Follicle/pathology , Hair Follicle/physiopathology , Humans , Immunity, Cellular , Macrophages/immunology , Major Histocompatibility Complex/immunology , Mast Cells/immunology , T-Lymphocytes/immunology
14.
J Investig Dermatol Symp Proc ; 4(3): 272-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10674380

ABSTRACT

Hair follicle (HF) morphogenesis and cycling are characterized by a tightly controlled balance of proliferation, differentiation and apoptosis. The members of the bcl-2 family of proto-oncogenes are important key players in the apoptosis control machinery of most cell types. Bcl-2, an apoptosis inhibitor, and Bax, an apoptosis promoter, show tightly regulated, hair cycle-dependent expression patterns: during catagen, the distal ORS of the HF remains strongly positive for Bcl-2 and Bax; in contrast, the proximal epithelial part of the HF loses most Bcl-2 expression while it remains strongly positive for Bax. In Bcl-2 null mice, skin becomes markedly hypopigmented during the first postnatal anagen probably due to increased melanocyte apoptosis. Reportedly, these mice also show a retardation of the first anagen development after birth. Transgenic mice overexpressing Bcl-2 under the control of the keratin-1 promoter display multifocal epidermal hyperplasia and aberrant expression of keratin-6, while alterations of HF cycling have not been investigated. Surprisingly, Bcl-2 overexpression under the control of the keratin-14 promoter leads to accelerated catagen progression and increased chemotherapy-induced apoptosis, HF dystrophy and alopecia. Transgenic mice overexpressing Bcl-X(L), another anti-apoptotic bcl-2 family member, under the control of the K14 promoter, reportedly also display accelerated catagen development. These and other Bcl-2 transgenic and null mice are now available to further dissect the as yet unclear, and likely complex, role of Bcl-2 in HF growth and pigmentation.


Subject(s)
Apoptosis/physiology , Genes, bcl-2 , Hair Follicle/pathology , Hair Follicle/physiology , Animals , Gene Expression Regulation , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/physiology
15.
J Investig Dermatol Symp Proc ; 4(3): 338-45, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10674393

ABSTRACT

The hair follicle (HF) is the only mammalian organ that undergoes life-long, cyclic transformations from long stages of growth (anagen), via rapid, apoptosis-driven organ involution (catagen) to a stage of relative "resting" (telogen). The controls that underlie these transformations clearly reside in and/or around the HF itself, and are likely to reflect - essentially autonomous, yet highly manipulable - changes in the local signalling milieu of e.g., hair growth-modulatory growth factors, cytokines, hormones and adhesion molecules. Yet the molecular nature and organization of the "hair cycle clock" (HCC) that drives these cyclic switches in the local signalling milieu remain obscure, and there is not even a fully satisfactory theory of hair cycle control. Since deciphering of the HCC is of paramount clinical importance, and since corresponding working hypotheses are badly needed to guide the design of more incisive experiments that identify the elusive central "oscillator" mechanism behind the HCC, we discuss basic requirements any convincing HCC theory should meet. After arguing that at least four distinct timing devices underlie HF chronobiology ("morphogenesis clock", "cycling inducer", "desynchronizer", and the actual HCC), previously proposed HCC theories are briefly and critically reviewed. In the light of intriguing regulatory similarities between the HCC and the cell cycle machinery, we suggest here that the HCC may be driven by autonomous, cell cycle-coupled secretory activities of the HF mesenchyme, namely by changes in the G0/G1-associated secretion of "papilla morphogens" by dermal papilla fibroblasts. Hopefully, this provocative hypothesis will encourage the proposition of novel, comprehensive HCC theories.


Subject(s)
Biological Clocks , Hair Follicle/cytology , Hair Follicle/physiology , Animals , Cell Differentiation , Cell Division , Humans , Signal Transduction
16.
J Histochem Cytochem ; 46(12): 1401-10, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9815282

ABSTRACT

Hair follicle development, growth (anagen), and regression (catagen) largely result from bidirectional epithelial-mesenchymal interactions whose molecular basis is still unclear. Because adhesion molecules are critically involved in pattern formation and because the fundamental importance of neural cell adhesion molecule (NCAM) for feather development has been demonstrated, we studied the protein expression patterns of NCAM during hair follicle development and regression in the C57BL/6 mouse model. During murine hair follicle development, NCAM immunoreactivity (IR) was first detected on epithelial hair placodes and later on selected keratinocytes in the distal outer root sheath. Mesenchymal NCAM immunoreactivity (IR) was noted on fibroblasts of the future dermal papilla (DP) and the perifollicular connective tissue sheath. Fetal hair follicle elongation coincided with strong, ubiquitous dermal NCAM IR, which remained strong until the follicles entered into their first neonatal catagen. At this time, the strong interfollicular dermal NCAM IR decreased substantially. During consecutive hair cycles, mesenchymal NCAM IR was seen exclusively on DP and perifollicular connective tissue sheath fibroblasts and on the trailing cells of regressing catagen hair follicles. These highly restricted and developmentally controlled expression patterns suggest an important role for NCAM in hair follicle topobiology during morphogenesis and cyclic remodeling of this miniorgan.


Subject(s)
Hair Follicle/growth & development , Hair Follicle/metabolism , Neural Cell Adhesion Molecules/metabolism , Aging , Animals , Female , Hair Follicle/embryology , Immunohistochemistry , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Periodicity , Skin/embryology , Skin/growth & development , Skin/metabolism
17.
J Invest Dermatol ; 111(1): 7-18, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9665380

ABSTRACT

In this immunohistomorphometric study, we have defined basic characteristics of the hair follicle (HF) immune system during follicle morphogenesis and cycling in C57BL/6 mice, in relation to the skin immune system. Langerhans cells and gammadelta T cell receptor immunoreactive lymphocytes were the predominant intraepithelial hematopoietic cells in neonatal mouse skin. After their numeric increase in the epidermis, these cells migrated into the HF, although only when follicle morphogenesis was almost completed. In contrast to Langerhans cells, gammadelta T cell receptor immunoreactive lymphocytes entered the HF only via the epidermis. Throughout HF morphogenesis and cycling, both cell types remained strikingly restricted to the distal outer root sheath. On extremely rare occasions, CD4+ or CD8+ alphabetaTC were detected within the HF epithelium or the sebaceous gland. Major histocompatibility complex class II+, MAC-1+ cells of macrophage phenotype and numerous mast cells appeared very early on during HF development in the perifollicular dermis, and the percentage of degranulated mast cells significantly increased during the initiation of synchronized HF cycling (first catagen). During both depilation- and cyclosporine A-induced HF cycling, the numbers of intrafollicular Langerhans cells, gammadelta T cell receptor immunoreactive lymphocytes, and perifollicular dermal macrophages fluctuated significantly. Yet, no numeric increase of perifollicular macrophages was detectable during HF regression, questioning their proposed role in catagen induction. In summary, the HF immune system is generated fairly late during follicle development, shows striking differences to the extrafollicular skin immune system, and undergoes substantial hair cycle-associated remodeling. In addition, synchronized HF cycling is accompanied by profound alterations of the skin immune system.


Subject(s)
Hair Follicle/immunology , Immune System/physiology , Integrin alpha Chains , Animals , Animals, Newborn/immunology , Antigens, CD/analysis , Cadherins/analysis , Dendritic Cells/physiology , Histocompatibility Antigens Class II/analysis , Langerhans Cells/physiology , Lymphocyte Count , Macrophages/physiology , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/analysis
18.
J Invest Dermatol ; 110(6): 902-7, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9620297

ABSTRACT

Mutation of the hairless (hr) gene in mice causes severe abnormalities during the first hair follicle regression (catagen), resulting in complete baldness. Here, we further characterize how hairlessness develops in HRS/J hairless mouse skin (hr) by histology, histochemistry, immunohistology, and in situ hybridization. We show that, in hr skin, only two defined epithelial cell populations in the distal outer root sheath (ORS) retain their integrity, whereas the rest of the ORS disintegrates. The surviving distal ORS forms the characteristic utriculi, whereas the remnants of the bulge get isolated from other epithelial compartments, but retain the capacity to proliferate and to produce either columnar epithelial outgrowths or selected dermal cysts. Normal dermal papilla structures get lost during the development of hairlessness. Based on the patterns of keratin 17 mRNA and neural cell adhesion molecule antigen expression, and on the distribution of alkaline phosphatase activity, we propose that dermal cysts in hr skin arise from (i) the central ORS, (ii) bulge-derived cells, or (iii) the disintegrating proximal ORS under the influence of dermal papilla remnants. The hr mutation seems to disrupt the integrity of key functional tissue units in the hair follicle, possibly due to a dysregulation of normal, catagen-associated apoptosis and/or an impairment of cell adhesion, whereas the distal follicle epithelium (including its stem cell region) seems to be largely protected from this. Thus, hairless mice offer a unique model for dissecting the as yet obscure functional properties of the hr gene product in maintaining follicle integrity during normal catagen.


Subject(s)
Hair/pathology , Mice, Hairless/genetics , Skin Diseases/etiology , Alkaline Phosphatase/metabolism , Animals , Female , Gene Expression/genetics , Immunohistochemistry , In Situ Hybridization , Keratins/genetics , Ki-67 Antigen/genetics , Mice , Mice, Inbred C57BL , Neural Cell Adhesion Molecules/genetics , Phenotype , RNA, Messenger/analysis , RNA, Messenger/genetics , Skin/chemistry , Skin/enzymology , Skin/pathology , Skin Diseases/genetics , Skin Diseases/pathology
19.
J Histochem Cytochem ; 46(3): 361-70, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9487118

ABSTRACT

In back skin sections from adolescent C57BL/6 mice, regularly distributed, perifollicular inflammatory cell clusters (PICC) were found located around the distal noncycling portion of about 2% of all hair follicles examined. The PICC and the affected hair follicles were characterized during spontaneously developed or induced hair cycle stages, using antibodies against MHC Class II, F4/80, ER-MP23, NLDC 145, CD4, CD8, gammadeltaTCR, IL-1 receptor, and ICAM-1. PICC consisted predominantly of macrophages (MAC), accompanied by a few CD4+ cells, whereas gammadeltaTCR+ and CD8+ cells were absent. During anagen and catagen, some of the PICC+ hair follicles showed variable degenerative phenomena reminiscent of scarring alopecia: thickened basement membrane, ectopic MHC II expression, MAC infiltration into the follicle epithelium, and signs of keratinocyte apoptosis. Loss of distal outer root sheath keratinocytes was detected in 10% of PICC+ hair follicles (0.2% of all hair follicles). Because PICC were located in the vicinity of the bulge region, MAC-dependent damage to follicle stem cells might eventually lead to follicle degeneration. These perifollicular MAC clusters around selected hair follicles may indicate the existence of a physiological program of MAC-dependent controlled follicle degeneration by which damaged or malfunctioning follicles are removed by programmed organ deletion (POD).


Subject(s)
Hair Follicle/growth & development , Hair Follicle/immunology , Macrophages/cytology , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Female , Genes, MHC Class II , Histocytochemistry , Immunohistochemistry , Intercellular Adhesion Molecule-1/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1 Type I
20.
Cell Immunol ; 178(2): 172-9, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9225008

ABSTRACT

The development of murine contact hypersensitivity is influenced by hair follicle cycling. Here, we have examined hair cycle-associated fluctuations of murine contact photosensitivity (CPS) to tetrachlorosalicylanilide (TCSA) and its immunologic mechanism(s). When the CPS outcome was monitored in correlation with their spontaneous, synchronized hair cycling, mice aged 8 and 14 weeks, with most of their hair follicles in telogen, exhibited strong CPS responses, whereas 4-, 11-, and 16-week-old mice with a predominance of anagen follicles in a large area of their integument exhibited lower responses. This suggests that the development of CPS is inhibited in mice with anagen hair follicles. Antigen-specific, T-cell receptor V beta 7+ suppressor T cells, which are recognized to down-regulate the CPS response to TCSA, were not generated in sensitized anagen mice. Culture supernatants of epidermal cells derived from mice with anagen hair follicles contained factor(s) that suppress in vivo the development of CPS. It was found that levels of mRNA for tumor necrosis factor alpha (TNF alpha) were markedly decreased in epidermal cells from early anagen to telogen mice, whereas message for IL-1 receptor antagonist (IL-1ra) was transcribed increasingly during this hair cycling. These findings suggest that altered keratinocyte production of these cytokines is involved in mediating the anagen-associated depression of CPS.


Subject(s)
Dermatitis, Contact/immunology , Hair Follicle/immunology , Hypersensitivity, Delayed/immunology , Age Factors , Animals , Cytokines/physiology , Gene Expression , Interleukin-1/physiology , Interleukin-10/physiology , Keratinocytes/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Salicylanilides/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/genetics , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...