Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Am Acad Dermatol ; 90(4): 790-797, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37992812

ABSTRACT

BACKGROUND: Anti-p200 pemphigoid is a subepidermal autoimmune blistering disease (AIBD) characterized by autoantibodies against a 200 kDa protein. Laminin γ1 has been described as target antigen in 70% to 90% of patients. No diagnostic assay is widely available for anti-p200 pemphigoid, which might be due to the unclear pathogenic relevance of anti-laminin γ1 autoantibodies. OBJECTIVE: To identify a target antigen with higher clinical and diagnostic relevance. METHODS: Immunoprecipitation, mass spectrometry, and immunoblotting were employed for analysis of skin extracts and sera of patients with anti-p200 pemphigoid (n = 60), other AIBD (n = 33), and healthy blood donors (n = 29). To localize the new antigen in skin, cultured keratinocytes and fibroblasts, quantitative real-time polymerase chain reaction and immunofluorescence microscopy were performed. RESULTS: Laminin ß4 was identified as target antigen of anti-p200 pemphigoid in all analyzed patients. It was located at the level of the basement membrane zone of the skin with predominant expression in keratinocytes. LIMITATIONS: A higher number of sera needs to be tested to verify that laminin ß4 is the diagnostically relevant antigen of anti-p200 pemphigoid. CONCLUSION: The identification of laminin ß4 as an additional target antigen in anti-p200 pemphigoid will allow its differentiation from other AIBD and as such, improve the management of these rare disorders.


Subject(s)
Pemphigoid, Bullous , Humans , Autoantibodies , Autoantigens , Basement Membrane , Blister , Laminin , Giardia
2.
J Invest Dermatol ; 141(11): 2602-2610.e3, 2021 11.
Article in English | MEDLINE | ID: mdl-33965403

ABSTRACT

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.


Subject(s)
Cell Adhesion Molecules/physiology , Homeostasis/physiology , Keratinocytes/cytology , Actin Cytoskeleton/physiology , Alarmins/physiology , Animals , Blister/etiology , Cell Differentiation , Epidermis/pathology , Keratins/analysis , Mice , Receptor, Fibroblast Growth Factor, Type 1/analysis , Kalinin
3.
Exp Dermatol ; 30(1): 17-24, 2021 01.
Article in English | MEDLINE | ID: mdl-33205478

ABSTRACT

The basement membrane at the dermal-epidermal junction keeps the epidermis attached to the dermis. This anatomical barrier is made up of four categories of extracellular matrix proteins: collagen IV, laminin, nidogen and perlecan. These proteins are precisely arranged in a well-defined architecture through specific interactions between the structural domains of the individual components. Some of the molecular constituents are provided by both fibroblasts and keratinocytes, while others are synthesized exclusively by fibroblasts or keratinocytes. It remains to be determined how the components from the fibroblasts are targeted to the dermal-epidermal junction and correctly organized and integrated with the proteins from the adjacent keratinocytes to form the basement membrane.


Subject(s)
Basement Membrane/metabolism , Dermis/metabolism , Epidermis/metabolism , Laminin/metabolism , Animals , Basement Membrane/anatomy & histology , Fibroblasts/metabolism , Humans , Keratinocytes/metabolism , Laminin/chemistry , Molecular Structure , Protein Isoforms/metabolism
4.
FASEB J ; 34(9): 12040-12052, 2020 09.
Article in English | MEDLINE | ID: mdl-32716577

ABSTRACT

Although collagens are the most abundant proteins implicated in various disease pathways, essential mechanisms required for their proper folding and assembly are poorly understood. Heat-shock protein 47 (HSP47), an ER-resident chaperone, was mainly reported to fulfill key functions in folding and secretion of fibrillar collagens by stabilizing pro-collagen triple-helices. In this study, we demonstrate unique functions of HSP47 for different collagen subfamilies. Our results show that HSP47 binds to the N-terminal region of procollagen I and is essential for its secretion. However, HSP47 ablation does not majorly impact collagen VI secretion, but its lateral assembly. Moreover, specific ablation of Hsp47 in murine keratinocytes revealed a new role for the transmembrane collagen XVII triple-helix formation. Incompletely folded collagen XVII C-termini protruding from isolated HSP47 null keratinocyte membrane vesicles could be fully restored upon the application of recombinant HSP47. Thus, our study expands the current view regarding the client repertoire and function of HSP47, as well as emphasizes its importance for transmembrane collagen folding.


Subject(s)
HSP47 Heat-Shock Proteins/metabolism , Keratinocytes/metabolism , Procollagen/metabolism , Protein Folding , Animals , HSP47 Heat-Shock Proteins/genetics , Mice , Procollagen/genetics
5.
Methods Cell Biol ; 143: 187-205, 2018.
Article in English | MEDLINE | ID: mdl-29310778

ABSTRACT

Laminins are large glycoproteins forming structural and signaling networks with two major physiological roles: one role crucial for the formation and stability of basement membranes and the other role, as crucial as the first, in cell anchorage and signaling. Laminins come in several flavors as 16 different isoforms are known, each with both common and unique functions. Here the most current techniques for purification and identification of laminins in tissues and cultivated cells as well as for testing the cell adhesion-promoting activity of laminins will be described.


Subject(s)
Cell Adhesion , Cell Culture Techniques/methods , Laminin/isolation & purification , Animals , Basement Membrane/chemistry , Cell Culture Techniques/instrumentation , Cells, Cultured , Extracellular Matrix/chemistry , Humans , Laminin/analysis , Laminin/physiology
6.
J Invest Dermatol ; 137(2): 332-340, 2017 02.
Article in English | MEDLINE | ID: mdl-27729280

ABSTRACT

Genetic, clinical, and biochemical studies have shown that integrity of the dermal-epidermal junction requires a particular subset of laminins, that is, those containing the α3 chain encoded by the Lama3 gene. Inherited mutations in the human gene or introduction of constitutive mutations in the mouse gene prevent expression of these laminins, causing junctional epidermolysis bullosa, a very severe, often lethal disorder characterized by detachment of the epidermis from the dermis. This has precluded in vivo functional analysis of α3 chain-containing laminins, and it is still unknown whether and how they contribute to adult skin homeostasis. To address this question, we have disrupted the Lama3 gene in basal keratinocytes of adult mice. This led to the gradual disappearance of α3 chain-containing laminins along the dermal-epidermal junction and formation of subepidermal blisters like in congenital junctional epidermis bullosa. The mice lose their nails and have bullae and erosions on the footpads. Because the blistering is restricted to the interfollicular epidermis, the animals do not lose the epidermis and are viable. There is abundant and scattered deposition of collagen VII on the dermal side of the blisters, inflammation, and development of skin fibrosis with extensive accumulation of interstitial and microfibrillar collagens.


Subject(s)
Dermatitis/etiology , Laminin/physiology , Skin/pathology , Animals , Collagen Type VII/metabolism , Fibrosis , Laminin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/metabolism
7.
Nat Commun ; 6: 8198, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26349061

ABSTRACT

Stem cells reside in specialized niches that are critical for their function. Quiescent hair follicle stem cells (HFSCs) are confined within the bulge niche, but how the molecular composition of the niche regulates stem cell behaviour is poorly understood. Here we show that integrin-linked kinase (ILK) is a key regulator of the bulge extracellular matrix microenvironment, thereby governing the activation and maintenance of HFSCs. ILK mediates deposition of inverse laminin (LN)-332 and LN-511 gradients within the basement membrane (BM) wrapping the hair follicles. The precise BM composition tunes activities of Wnt and transforming growth factor-ß pathways and subsequently regulates HFSC activation. Notably, reconstituting an optimal LN microenvironment restores the altered signalling in ILK-deficient cells. Aberrant stem cell activation in ILK-deficient epidermis leads to increased replicative stress, predisposing the tissue to carcinogenesis. Overall, our findings uncover a critical role for the BM niche in regulating stem cell activation and thereby skin homeostasis.


Subject(s)
Extracellular Matrix/genetics , Hair Follicle/cytology , Keratinocytes/metabolism , Protein Serine-Threonine Kinases/genetics , Stem Cell Niche/genetics , Stem Cells/metabolism , Animals , Basement Membrane , Blotting, Western , Carcinogens/pharmacology , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Proliferation , Electrophoresis, Polyacrylamide Gel , Epidermal Cells , Extracellular Matrix/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockout Techniques , Immunohistochemistry , Laminin/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Skin/drug effects , Stem Cells/cytology , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor beta , Wnt Signaling Pathway , Kalinin
8.
Anticancer Agents Med Chem ; 14(4): 578-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24628265

ABSTRACT

Integrin-dependent adhesion of tumor cells to extracellular matrix proteins provides anchorage-dependent protection from cell death. In the present investigation we aimed to understand whether and how the paradigmatic membrane-targeted synthetic phospholipid analog erufosine is relevant for tumor cell adhesion to extracellular matrix proteins, cell survival and migration. The antineoplastic action of erufosine was analyzed with glioblastoma and prostate cancer cells adhering to fibronectin or collagen I using proliferation, adhesion and migration assays. The composition of adhesion contacts containing activated ß1 integrins was studied using immunofluorescence. The importance of ß1 integrins for the observed effects was analyzed in fibroblasts proficient or deficient in ß1 integrin expression. Adhesion to collagen I and fibronectin increased the death threshold in serum-deprived tumor cells. Moreover, ß1 integrin-deficient cells were more sensitive to erufosine-treatment compared to ß1 integrin proficient cells suggesting a role of ß1 integrins for matrix-mediated death resistance. Most importantly, erufosine disturbed the maturation of the cell adhesion complexes containing paxillin, activated ß1 integrins and phosphorylated FAK, leading to a reduction of survival signals and inhibition of tumor cell adhesion and migration. These findings suggest that membrane-targeted synthetic phospholipids analogs may be of value for counteracting matrix-mediated treatment resistance in combined treatment approaches with radiotherapy or chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/drug effects , Extracellular Matrix/drug effects , Organophosphates/pharmacology , Quaternary Ammonium Compounds/pharmacology , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , Collagen Type I/physiology , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins/physiology , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioblastoma , Humans , Integrin beta1/metabolism , Male , Mice , Paxillin/metabolism , Phosphorylation , Prostatic Neoplasms
9.
Cell Adh Migr ; 7(1): 48-55, 2013.
Article in English | MEDLINE | ID: mdl-23263632

ABSTRACT

Laminins are large molecular weight glycoproteins constituted by the assembly of three disulfide-linked polypeptides, the α, ß and γ chains. The human genome encodes 11 genetically distinct laminin chains. Structurally, laminin chains differ by the number, size and organization of a few constitutive domains, endowing the various members of the laminin family with common and unique important functions. In particular, laminins are indispensable building blocks for cellular networks physically bridging the intracellular and extracellular compartments and relaying signals critical for cellular behavior, and for extracellular polymers determining the architecture and the physiology of basement membranes.


Subject(s)
Basement Membrane/metabolism , Laminin/metabolism , Signal Transduction , Animals , Basement Membrane/cytology , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Humans , Laminin/genetics , Molecular Weight , Protein Conformation , Protein Folding , Protein Interaction Mapping , Protein Structure, Tertiary , Receptors, Laminin/metabolism
10.
Am J Pathol ; 180(4): 1581-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326752

ABSTRACT

Kindlin-1 is an adaptor protein that is expressed by most epithelial cells and has been implicated in integrin bidirectional signaling. Mutations in the gene encoding kindlin-1 are associated with Kindler syndrome, a recessively inherited disorder that is characterized by fragile skin. Functionally, a loss of kindlin-1 impairs the adhesion of basal keratinocytes to the extracellular matrix both in vivo and in vitro. In this study, we show that the phenotype of mutant keratinocytes deficient in kindlin-1 is characterized by the modification of the cortical actin network and increased plasticity of the plasma membrane. At the molecular level, expression of several proteins associated with an epithelial phenotype, such as α6ß4 integrin, collagen XVII, E-cadherin, and desmoglein-3, is strongly reduced, whereas, surprisingly, laminin 332 is synthesized in larger amounts than in control keratinocytes. In contrast, mesenchymal markers such as vimentin and fibronectin are increased in keratinocytes lacking kindlin-1. The switch in cell plasticity and protein expression was confirmed by siRNA-mediated down-regulation of kindlin-1 in HaCaT epithelial cells. Furthermore, there was up-regulation of matrix metalloproteinases and pro-inflammatory cytokines in kindlin-1-deficient keratinocytes. These results provide new insights into the pathogenic mechanisms that take place in Kindler syndrome. Moreover, the constellation of molecular defects associated with the loss of kindlin-1 may explain the higher incidence of skin cancer observed in patients affected with this disorder.


Subject(s)
Blister/metabolism , Epidermolysis Bullosa/metabolism , Keratinocytes/metabolism , Membrane Proteins/deficiency , Neoplasm Proteins/deficiency , Periodontal Diseases/metabolism , Photosensitivity Disorders/metabolism , Actin Cytoskeleton/metabolism , Blister/genetics , Blister/pathology , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Shape/genetics , Cell Shape/physiology , Cells, Cultured , Cytokines/metabolism , Down-Regulation/physiology , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/pathology , Epithelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Integrin beta4/metabolism , Keratinocytes/pathology , Keratinocytes/physiology , Matrix Metalloproteinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Phase-Contrast , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Periodontal Diseases/genetics , Periodontal Diseases/pathology , Phenotype , Photosensitivity Disorders/genetics , Photosensitivity Disorders/pathology , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Stem Cells/metabolism , Transcription Factors/metabolism
11.
Exp Dermatol ; 20(8): 689-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21615511

ABSTRACT

The current understanding of the role of extracellular matrix proteins is mainly based on their structural properties and their assembly into complex networks. The multiplicity of interactions between cells, cytokines and growth factors within the networks determines functional units dictating the biophysical properties of tissues. This review focuses on the understanding how alterations in the genes, modifying enzymes or biological functions of extracellular matrix molecules, lead to inborn or acquired skin disorders. Analysis of the disease mechanisms provides the basis for the emerging concept that not solely structural defects of single extracellular matrix proteins are at fault, but rather that the functional unit as a whole is not working properly, causing similar clinical symptoms although the causative genes are entirely different. The understanding of these disease-causing pathways has already led to surprising new therapeutic developments applied to rare inborn disorders. They now permit to design new concepts for the treatment of more common diseases associated with the accumulation of connective tissue and alterations of the biomechanical properties of the extracellular matrix.


Subject(s)
Dermis/physiology , Extracellular Matrix Proteins/physiology , Extracellular Matrix/physiology , Collagen/physiology , Humans , Signal Transduction/physiology , Skin Diseases/physiopathology
12.
Eur J Cell Biol ; 90(1): 1-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20971525

ABSTRACT

Spatial and temporal expression of laminin isoforms is assumed to provide specific local information to neighboring cells. Here, we report the remarkably selective presence of LM-111 at the very tip of hair follicles where LM-332 is absent, suggesting that epithelial cells lining the dermal-epidermal junction at this location may receive different signals from the two laminins. This hypothesis was tested in vitro by characterizing with functional and molecular assays the comportment of keratinocytes exposed to LM-111 and LM-332. The two laminins induced morphologically distinct focal adhesions, and LM-332, but not LM-111, elicited persistent migration of keratinocytes. The different impact on cellular behavior was associated with distinct activation patterns of Rho GTPases and other signaling intermediates. In particular, while LM-111 triggered a robust activation of Cdc42, LM-332 provoked a strong and sustained activation of FAK. Interestingly, activation of Rac1 was necessary but not sufficient to promote migration because there was no directed migration on LM-111 despite Rac1 activation. In contrast, RhoA antagonized directional migration, since silencing of RhoA by RNA interference boosted unidirectional migration on LM-332. Molecular analysis of the role of RhoA strongly suggested that the mechanisms involve disassembly of cell-cell contacts, loss of the cortical actin network, mobilization of α6ß4 integrin out of stable adhesions, and displacement of the integrin from its association with the insoluble pool of intermediate filaments.


Subject(s)
Cell Movement , Keratinocytes/physiology , Laminin/physiology , rhoA GTP-Binding Protein/metabolism , Cell Line , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression , Hair Follicle/metabolism , Humans , Immunoblotting , Integrins/metabolism , Keratinocytes/metabolism , Laminin/genetics , Microscopy , RNA Interference , Signal Transduction , Time-Lapse Imaging , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
13.
Wound Repair Regen ; 18(3): 325-34, 2010.
Article in English | MEDLINE | ID: mdl-20412552

ABSTRACT

Two integrins, alpha3beta1 and alpha6beta4, are high-affinity receptors for laminin 332, the major laminin isoform of the dermal-epidermal junction, although they are thought to have different functions. Biological and genetic studies have firmly established that the alpha6beta4 integrin is indispensable for the stable anchorage of the epidermis to the underlying dermis. In contrast, the alpha3beta1 integrin is thought to be important for cell migration, although the issue is controversial, and both positive and negative effects have been reported. To address the function of alpha3beta1 integrin, we used small interfering RNA to down-regulate the alpha3 subunit in human keratinocytes. The resulting phenotype indicates that lack of alpha3beta1 integrin compromises intercellular adhesion and collective migration, while it enhances single cell migration with a concomitant increase of both focal adhesion kinase and extracellular signal-regulated kinase. In addition, down-regulation of integrin alpha3 subunit results in an increased expression of fibronectin and precursor laminin 332, two extracellular matrix proteins known to be up-regulated during wound healing. Thus, down-regulation of alpha3beta1 integrin recapitulates crucial events governing keratinocyte migration associated with wound healing and tissue repair.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Integrin alpha3beta1/physiology , Keratinocytes/metabolism , Wound Healing/physiology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Fibronectins/metabolism , Humans , Kalinin
14.
Am J Pathol ; 175(4): 1442-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19762715

ABSTRACT

Kindlin-1 is an epithelial-specific member of the novel kindlin protein family, which are regulators of integrin functions. Mutations in the gene that encodes Kindlin-1, FERMT1 (KIND1), cause the Kindler syndrome (KS), a human disorder characterized by mucocutaneous fragility, progressive skin atrophy, ulcerative colitis, photosensitivity, and propensity to skin cancer. Our previous studies indicated that loss of kindlin-1 resulted in abnormalities associated with integrin functions, such as adhesion, proliferation, polarization, and motility of epidermal cells. Here, we disclosed novel FERMT1 mutations in KS and used them, in combination with small-interfering RNA, protein, and imaging studies, to uncover new functions for kindlin-1 in keratinocytes and to discern the molecular pathology of KS. We show that kindlin-1 forms molecular complexes with beta1 integrin, alpha-actinin, migfilin, and focal adhesion kinase and regulates cell shape and migration by controlling lamellipodia formation. Kindlin-1 governs these processes by signaling via Rho family GTPases, and it is required to maintain the pool of GTP-bound, active Rac1, RhoA and Cdc42, and the phosphorylation of their downstream effectors p21-activated kinase 1, LIM kinase, and cofilin. Loss of these kindlin-1 functions forms the biological basis for the epithelial cell fragility and atrophy in the pathology of KS.


Subject(s)
Keratinocytes/enzymology , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Pseudopodia/enzymology , rho GTP-Binding Proteins/metabolism , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Adult , Cell Line, Transformed , Cell Movement , Cell Shape , Child , Enzyme Activation , Focal Adhesions/enzymology , Guanosine Triphosphate/metabolism , Humans , Keratinocytes/pathology , Middle Aged , Models, Biological , Mucous Membrane/abnormalities , Mucous Membrane/pathology , Phenotype , Phosphorylation , Protein Binding , RNA, Small Interfering/metabolism , Skin Abnormalities/enzymology , Skin Abnormalities/pathology , Syndrome
15.
J Exp Med ; 204(11): 2615-27, 2007 Oct 29.
Article in English | MEDLINE | ID: mdl-17923499

ABSTRACT

B cell homeostasis is regulated by multiple signaling processes, including nuclear factor-kappaB (NF-kappaB), BAFF-, and B cell receptor signaling. Conditional disruption of genes involved in these pathways has shed light on the mechanisms governing signaling from the cell surface to the nucleus. We describe a novel mouse strain that expresses solely and excessively a naturally occurring splice variant of CYLD (CYLD(ex7/8) mice), which is a deubiquitinating enzyme that is integral to NF-kappaB signaling. This shorter CYLD protein lacks the TRAF2 and NEMO binding sites present in full-length CYLD. A dramatic expansion of mature B lymphocyte populations in all peripheral lymphoid organs occurs in this strain. The B lymphocytes themselves exhibit prolonged survival and manifest a variety of signaling disarrangements that do not occur in mice with a complete deletion of CYLD. Although both the full-length and the mutant CYLD are able to interact with Bcl-3, a predominant nuclear accumulation of Bcl-3 occurs in the CYLD mutant B cells. More dramatic, however, is the accumulation of the NF-kappaB proteins p100 and RelB in CYLD(ex7/8) B cells, which, presumably in combination with nuclear Bcl-3, results in increased levels of Bcl-2 expression. These findings suggest that CYLD can both positively and negatively regulate signal transduction and homeostasis of B cells in vivo, depending on the expression of CYLD splice variants.


Subject(s)
B-Lymphocytes/immunology , Cysteine Endopeptidases/genetics , Alternative Splicing , Animals , Deubiquitinating Enzyme CYLD , Embryonic Stem Cells/immunology , Embryonic Stem Cells/physiology , Exons , Fibroblasts/physiology , Genetic Variation , Homeostasis , Mice , Protein Biosynthesis , Sequence Deletion , Signal Transduction
16.
Eur J Cell Biol ; 86(11-12): 719-30, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17689829

ABSTRACT

CD1a(pos) dendritic cells (DCs) and Langerhans cells (LCs) are highly specialized antigen-presenting cells mainly localized in the skin. Various cells have been identified as precursors of cutaneous DCs, but the definitive precursor subpopulations remain to be defined and characterized in detail. In this study, DCs were generated in vitro from monocytes (monocyte-derived DCs, MoDCs) and from CD34(pos) stem cells (CD34(pos) cell-derived DCs, CD34DCs). By virtue of their CD14 and CD1a expression, four CD34DC subpopulations were characterized while MoDCs contain three different subpopulations. Of these, CD14-expressing cells are considered to be precursors of fully differentiated DCs, which themselves are CD14(neg)CD1a(pos). Both, MoDCs and CD34DCs expressed the alpha integrins LFA-1, Mac-1, CR4, VLA-4, VLA-5 and the beta2 integrin CD18. CD34DCs and MoDCs were negative for VLA-3, whereas MoDCs, but not CD34DCs expressed VLA-6. Phenotypic and functional characterization of the cells generated herein at earlier time points revealed that DCs at day 3 of culture may reflect the in vivo situation more closely than at day 7. Adhesion of DC precursors to endothelial cells and to components of the extracellular matrix is a prerequisite for their migration towards the epidermis. To this end, we investigated adhesion of CD34DCs and MoDCs to components of the cutaneous extracellular matrix. Distinct DC subsets showed a differential binding pattern to proteins of the extracellular matrix. MoDCs and CD34DCs bound preferentially to laminin 332 via CD49f and to fibronectin via CD49e, but only weakly to laminin 111 or to collagens. While CD14(pos) cells preferentially bound to laminin 332, CD1a(pos) cells adhered to fibronectin. In summary, subpopulations of CD34DCs and MoDCs are phenotypically related to each other, but not identical and display differential binding to components of the extracellular matrix.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , Extracellular Matrix Proteins/metabolism , Antigens, CD34/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Humans , Integrins/metabolism , Phenotype , Protein Subunits/metabolism , Kalinin
17.
J Cell Biol ; 177(3): 501-13, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17485490

ABSTRACT

Integrin-linked kinase (ILK) links integrins to the actin cytoskeleton and is believed to phosphorylate several target proteins. We report that a keratinocyte-restricted deletion of the ILK gene leads to epidermal defects and hair loss. ILK-deficient epidermal keratinocytes exhibited a pronounced integrin-mediated adhesion defect leading to epidermal detachment and blister formation, disruption of the epidermal-dermal basement membrane, and the translocation of proliferating, integrin-expressing keratinocytes to suprabasal epidermal cell layers. The mutant hair follicles were capable of producing hair shaft and inner root sheath cells and contained stem cells and generated proliferating progenitor cells, which were impaired in their downward migration and hence accumulated in the outer root sheath and failed to replenish the hair matrix. In vitro studies with primary ILK-deficient keratinocytes attributed the migration defect to a reduced migration velocity and an impaired stabilization of the leading-edge lamellipodia, which compromised directional and persistent migration. We conclude that ILK plays important roles for epidermis and hair follicle morphogenesis by modulating integrin-mediated adhesion, actin reorganization, and plasma membrane dynamics in keratinocytes.


Subject(s)
Cell Movement , Hair Follicle/embryology , Keratinocytes/metabolism , Organogenesis , Protein Serine-Threonine Kinases/metabolism , Stem Cells/metabolism , Actins/genetics , Actins/metabolism , Animals , Basement Membrane/cytology , Basement Membrane/embryology , Blister/genetics , Blister/metabolism , Cell Adhesion/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Movement/genetics , Cell Proliferation , Dermis/cytology , Dermis/embryology , Female , Hair Follicle/cytology , Keratinocytes/cytology , Male , Mice , Mice, Transgenic , Organ Specificity/genetics , Organogenesis/genetics , Protein Serine-Threonine Kinases/deficiency , Pseudopodia/genetics , Pseudopodia/metabolism , Stem Cells/cytology
18.
J Investig Dermatol Symp Proc ; 11(1): 66-72, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17069012

ABSTRACT

The extracellular matrix (ECM) environment in connective tissues provides fibroblasts with a structural scaffold and modulates cell shape, but it also profoundly influences the fibroblast phenotype. Here we studied fibroblasts cultured in a three-dimensional network of native collagen, which was either mechanically stressed or relaxed. Mechanical load induces fibroblasts that synthesize abundant ECM and a characteristic array of cytokines/chemokines. This phenotype is reminiscent of late granulation tissue or scleroderma fibroblasts. By contrast, relaxed fibroblasts are characterized by induction of proteases and a subset of cytokines that does not overlap with that of mechanically stimulated cells. Thus, the biochemical composition and physical nature of the ECM exert powerful control over the phenotypes of fibroblasts, ranging from "synthetic" to "inflammatory" phenotypes. Interactions between fibroblasts and collagen fibrils are mostly mediated by a subset of beta 1 integrin receptors. Fibroblasts utilize alpha 1 beta 1, alpha 2 beta 1, and alpha 11 beta 1 integrins for establishing collagen contacts and transducing signals. In vitro assays and mouse genetics have demonstrated individual tasks served by each receptor, but also functional redundancy. Unraveling the integrated functions of fibroblasts, collagen integrin receptors, collagen fibrils, and mechanical tension will be important to understand the molecular mechanisms underlying tissue repair and fibrosis.


Subject(s)
Fibroblasts/physiology , Integrin alpha2beta1/physiology , Wound Healing/physiology , Animals , Chemokine CCL2/biosynthesis , Cyclooxygenase 2/physiology , Extracellular Matrix/metabolism , Humans , Mice , Phenotype , Scleroderma, Systemic/metabolism , Stress, Mechanical
19.
J Biol Chem ; 281(47): 36082-90, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17012746

ABSTRACT

A novel family of focal adhesion proteins, the kindlins, is involved in attachment of the actin cytoskeleton to the plasma membrane and in integrin-mediated cellular processes. Deficiency of kindlin-1, as a result of loss-of-function mutations in the KIND1 gene, causes Kindler syndrome, an autosomal recessive genodermatosis characterized by skin blistering, progressive skin atrophy, photosensitivity and, occasionally, carcinogenesis. Here we characterized authentic and recombinantly expressed kindlin-1 and show that it is localized in basal epidermal keratinocytes in a polar fashion, close to the cell surface facing the basement membrane, in the areas between the hemidesmosomes. We identified two forms of kindlin-1 in keratinocytes, with apparent molecular masses of 78 and 74 kDa, corresponding to phosphorylated and desphosphorylated forms of the protein. In kindlin-1-deficient skin, basal keratinocytes show multiple abnormalities: cell polarity is lost, proliferation is strongly reduced, and several cells undergo apoptosis. In vitro, deficiency of kindlin-1 in keratinocytes leads to strongly reduced cell proliferation, decreased adhesion, undirected motility, and intense protrusion activity of the plasma membrane. Taken together, these results show that kindlin-1 plays a role in keratinocyte adhesion, polarization, proliferation, and migration. It is involved in organization and anchorage of the actin cytoskeleton to integrin-associated signaling platforms.


Subject(s)
Epidermal Cells , Keratinocytes/cytology , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Chlorocebus aethiops , Humans , Keratinocytes/metabolism , Membrane Proteins/chemistry , Neoplasm Proteins/chemistry , Phosphoproteins/chemistry , Phosphorylation , Recombinant Proteins/chemistry , Signal Transduction
20.
Expert Rev Mol Med ; 8(24): 1-21, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-17040578

ABSTRACT

Epidermolysis bullosa (EB) and associated skin-fragility syndromes are a group of inherited skin diseases characterised by trauma-induced blistering of the skin and mucous membranes. Mutations in at least 14 distinct genes encoding molecular components of the epidermis or the dermal-epidermal junction (DEJ) can cause blistering skin diseases that differ by clinical presentation and severity of the symptoms. Despite great advances in discerning the genetic basis of this group of diseases, the molecular pathways leading to symptoms are not yet fully understood. Unravelling these pathways by molecular analysis of the structure and in vitro assessment of functional properties of the human proteins involved, combined with genetic models in lower organisms, should pave the way for specific cures for inherited skin fragility.


Subject(s)
Blister/genetics , Blister/therapy , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/therapy , Animals , Blister/metabolism , Blister/pathology , Dermis/pathology , Dermis/ultrastructure , Epidermis/pathology , Epidermis/ultrastructure , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/therapy , Humans , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...