Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Br J Dermatol ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38820176

ABSTRACT

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a blistering disease caused by mutations in the gene encoding type VII collagen (C7). RDEB is associated with fibrosis, which is responsible for severe complications. The phenotypic variability observed in RDEB siblings suggests that epigenetic modifications contribute to disease severity. Identifying epigenetic changes may help to uncover molecular mechanisms underlying RDEB pathogenesis and new therapeutic targets. OBJECTIVES: To investigate histone acetylation in RDEB skin and to explore histone deacetylase inhibitors (HDACis) as therapeutic molecules capable of counteracting fibrosis and disease progression in RDEB mice. METHODS: Acetylated histone levels were detected in human skin by immunofluorescence and in RDEB fibroblasts by ELISA. The effects of Givinostat and valproic acid (VPA) on RDEB fibroblast fibrotic behaviour were assessed by collagen-gel contraction assay, Western blot and immunocytofluorescence for α-smooth muscle actin, ELISA for released transforming growth factor-ß1 (TGF-ß1). RNA-seq was performed in HDACi- and vehicle-treated RDEB fibroblasts. VPA was systemically administered to RDEB mice, and effects on overt phenotype were monitored. Fibrosis was investigated in the skin using histological and immunofluorescence analyses. Eye and tongue defects were examined microscopically. Mass spectrometry proteomics was performed on skin protein extracts from VPA-treated RDEB and control mice. RESULTS: Histone acetylation decreases in RDEB skin and primary fibroblasts. RDEB fibroblasts treated with HDACis lowered fibrotic traits including contractility, TGF-ß1 release, and proliferation. VPA administration to RDEB mice mitigated severe manifestations affecting eyes and paws. These effects were associated with fibrosis inhibition. Proteomic analysis of mouse skin revealed that VPA almost normalised protein sets involved in protein synthesis and immune response, processes linked to the increased susceptibility to cancer and bacterial infections observed in RDEB patients. CONCLUSIONS: Dysregulated histone acetylation contributes to RDEB pathogenesis by facilitating the progression of fibrosis. Repurposing of HDACi could be considered for disease-modifying treatments of RDEB.

2.
Int J Mol Sci ; 21(4)2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32093196

ABSTRACT

Variably reduced expression of the basement membrane component laminin-332 (α3aß3γ2) causes junctional epidermolysis bullosa generalized intermediate (JEB-GI), a skin fragility disorder with an increased susceptibility to squamous cell carcinoma (SCC) development in adulthood. Laminin-332 is highly expressed in several types of epithelial tumors and is central to signaling pathways that promote SCC tumorigenesis. However, laminin-332 mutations and expression in individuals affected by JEB-GI and suffering from recurrent SCCs have been poorly characterized. We studied a JEB-GI patient who developed over a hundred primary cutaneous SCCs. Molecular analysis combined with gene expression studies in patient skin and primary keratinocytes revealed that the patient is a functional hemizygous for the p.Cys1171* mutant allele which is transcribed in a stable mRNA encoding for a ß3 chain shortened of the last two C-terminal amino acids (Cys1171-Lys1172). The lack of the Cys1171 residue involved in the C-terminal disulphide bond to γ2 chain did not prevent assembly, secretion, and proteolytic processing of the heterotrimeric molecule. Immunohistochemistry of SCC specimens revealed accumulation of mutant laminin-332 at the epithelial-stromal interface of invasive front. We conclude that the C-terminal disulphide bond is a structural element crucial for laminin-332 adhesion function in-vivo. By saving laminin-332 amount, processing, and signaling role the p.Cys1171* mutation may allow intrinsic pro-tumorigenic properties of the protein to be conveyed, thus contributing to invasiveness and recurrence of SCCs in this patient.


Subject(s)
Carcinoma, Squamous Cell , Cell Adhesion Molecules , Epidermolysis Bullosa , Mutation , Neoplasm Proteins , Skin Neoplasms , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Epidermolysis Bullosa/pathology , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Kalinin
3.
Am J Pathol ; 187(7): 1445-1453, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28460207

ABSTRACT

Individuals with recessive dystrophic epidermolysis bullosa (RDEB), a rare genetic skin disease, carry mutations in the COL7A1 gene that codes for type VII collagen, an extracellular matrix component of the basement membrane zone forming the anchoring fibrils. As a consequence, RDEB individuals manifest unremitting skin blistering that evolves into chronic wounds, inflammation, and fibrosis. These features play a central role in the development of more severe disease complications, such as mitten deformities of hands and feet and aggressive epithelial cancers. Despite being recognized as a central clinical issue for RDEB, wound healing impairment has been only marginally investigated. Recently, studies with disease mouse models started to shed light on the molecular mechanisms underlying the altered healing response of RDEB. In turn, alterations found in RDEB skin cell behavior fostered the understanding of mechanisms that may be responsible for defective skin repair. This review summarizes findings related to healing impairment in RDEB, and highlights therapeutic strategies for ameliorating healing.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Wound Healing/genetics , Animals , Blister , Cell Proliferation , Disease Models, Animal , Epidermolysis Bullosa Dystrophica/pathology , Epidermolysis Bullosa Dystrophica/therapy , Genes, Recessive/genetics , Humans , Inflammation , Mice , Mutation , Skin/pathology
4.
Hum Mol Genet ; 23(15): 3907-22, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24599399

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a genodermatosis characterized by fragile skin forming blisters that heal invariably with scars. It is due to mutations in the COL7A1 gene encoding type VII collagen, the major component of anchoring fibrils connecting the cutaneous basement membrane to the dermis. Identical COL7A1 mutations often result in inter- and intra-familial disease variability, suggesting that additional modifiers contribute to RDEB course. Here, we studied a monozygotic twin pair with RDEB presenting markedly different phenotypic manifestations, while expressing similar amounts of collagen VII. Genome-wide expression analysis in twins' fibroblasts showed differential expression of genes associated with TGF-ß pathway inhibition. In particular, decorin, a skin matrix component with anti-fibrotic properties, was found to be more expressed in the less affected twin. Accordingly, fibroblasts from the more affected sibling manifested a profibrotic and contractile phenotype characterized by enhanced α-smooth muscle actin and plasminogen activator inhibitor 1 expression, collagen I release and collagen lattice contraction. These cells also produced increased amounts of proinflammatory cytokines interleukin 6 and monocyte chemoattractant protein-1. Both TGF-ß canonical (Smads) and non-canonical (MAPKs) pathways were basally more activated in the fibroblasts of the more affected twin. The profibrotic behaviour of these fibroblasts was suppressed by decorin delivery to cells. Our data show that the amount of type VII collagen is not the only determinant of RDEB clinical severity, and indicate an involvement of TGF-ß pathways in modulating disease variability. Moreover, our findings identify decorin as a possible anti-fibrotic/inflammatory agent for RDEB therapeutic intervention.


Subject(s)
Epidermolysis Bullosa Dystrophica/genetics , Fibroblasts/metabolism , Genotype , Phenotype , Skin/metabolism , Transforming Growth Factor beta/genetics , Twins, Monozygotic/genetics , Actins/genetics , Actins/metabolism , Adult , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Collagen Type VII/genetics , Collagen Type VII/metabolism , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Fibroblasts/pathology , Gene Expression Regulation , Genes, Recessive , Genetic Heterogeneity , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Severity of Illness Index , Signal Transduction , Skin/pathology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
5.
Wound Repair Regen ; 21(4): 545-53, 2013.
Article in English | MEDLINE | ID: mdl-23627689

ABSTRACT

Adipose tissue-derived stem cells (ASCs) are gaining increasing consideration in tissue repair therapeutic application. Recent evidence indicates that ASCs enhance skin repair in animal models of impaired wound healing. To assess the therapeutic activity of autologous vs. allogeneic ASCs in the treatment of diabetic ulcers, we functionally characterized diabetic ASCs and investigated their potential to promote wound healing with respect to nondiabetic ones. Adipose tissue-derived cells from streptozotocin-induced type 1 diabetic mice were analyzed either freshly isolated as stromal vascular fraction (SVF), or following a single passage of culture (ASCs). Diabetic ASCs showed decreased proliferative potential and migration. Expression of surface markers was altered in diabetic SVF and cultured ASCs, with a reduction in stem cell marker-positive cells. ASCs from diabetic mice released lower amounts of hepatocyte growth factor, vascular endothelial growth factor (VEGF)-A, and insulin-like growth factor-1, growth factors playing important roles in skin repair. Accordingly, the supernatant of diabetic ASCs manifested reduced capability to promote keratinocyte and fibroblast proliferation and migration. Therapeutic potential of diabetic SVF administered to wounds of diabetic mice was blunted as compared with cells isolated from nondiabetic mice. Our data indicate that diabetes alters ASC intrinsic properties and impairs their function, thus affecting therapeutic potential in the autologous treatment for diabetic ulcers.


Subject(s)
Adipose Tissue/cytology , Diabetes Mellitus, Experimental/physiopathology , Stem Cells/physiology , Wound Healing/physiology , Animals , Cell Movement/physiology , Cell Proliferation , Diabetes Mellitus, Experimental/metabolism , Fibroblasts/physiology , Hepatocyte Growth Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Keratinocytes/physiology , Male , Mice , Stem Cells/metabolism , Stromal Cells , Vascular Endothelial Growth Factor A/metabolism
6.
FASEB J ; 25(3): 916-27, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21098725

ABSTRACT

IL-22 has a pathogenetic role in psoriasis, where it is responsible for the altered proliferation and differentiation of keratinocytes and induces inflammatory molecules. The IL-22-induced effects are mediated by STAT3, whose activity is proportional to acetylation in lysine (Lys)685 and phosphorylation in tyrosine (Tyr)705. Lys 685 acetylation of STAT3 is inhibited by sirtuin (SIRT)1, a class III deacetylase promoting keratinocyte differentiation. Due to the opposite effects of IL-22 and SIRT1, we investigated whether IL-22-induced effects in keratinocytes could be regulated by SIRT1 through control of STAT3. We found that SIRT1 opposes the IL-22-induced STAT3 activity by deacetylating STAT3 and reducing STAT3 Tyr705 phosphorylation. By controlling STAT3, SIRT1 also influences the IL-22-induced expression of molecules involved in proliferation and inflammation as well as proliferation and migration processes in cultured keratinocytes. Although SIRT1 levels were similar in keratinocytes of healthy individuals and patients with psoriasis, they were reduced in psoriatic skin lesions, with the lymphokine IFN-γ inhibiting SIRT1 expression. Concomitantly, IFN-γ enhanced basal acetylation of STAT3 and its phosphorylation induced by IL-22. In conclusion, STAT3-dependent IL-22 signaling and effects in keratinocytes are negatively regulated by SIRT1. In skin affected by psoriasis, SIRT1 is down-regulated by IFN-γ, which thus renders psoriatic keratinocytes more prone to respond to IL-22.


Subject(s)
Interleukins/metabolism , Keratinocytes/metabolism , Psoriasis/metabolism , STAT3 Transcription Factor/metabolism , Sirtuin 1/metabolism , Acetylation/drug effects , Adult , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Dermatitis/immunology , Dermatitis/metabolism , Histone Deacetylases/metabolism , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Keratinocytes/cytology , Keratinocytes/immunology , Phosphorylation/physiology , Protein Processing, Post-Translational/physiology , Psoriasis/immunology , RNA, Small Interfering , STAT3 Transcription Factor/genetics , Signal Transduction/immunology , Sirtuin 1/genetics , Interleukin-22
7.
J Cutan Pathol ; 39(9): 826-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22804631

ABSTRACT

BACKGROUND: Vascular endothelial growth factor-C (VEGF-C), a lymphatic vessel growth factor, has been involved in the formation of lymph nodal metastases in different tumor types. Early evidences indicate that VEGF-C expression in human primary melanoma could be predictive of lymph nodal metastases, whereas the role of lymphangiogenesis is still controversial. METHODS: By immunohistochemical analysis, we investigated VEGF-C or CC chemokine receptor 7 expression, together with the lymphatic and blood vessel network, in 36 patients with primary skin melanomas and metastases at the sentinel lymph node biopsy (SLN-positive), and 26 melanoma patients with negative SLN biopsy (SLN-negative). RESULTS: We found that VEGF-C expression in primary melanoma specimens was significantly associated with SLN-positive (p < 0.001), particularly in thin melanomas. An association between augmented peritumoral lymphatic vessel area and SLN-positive (p < 0.02) was also seen. Conversely, no association between either expression of the CC chemokine receptor 7 in the primary tumor, or intratumoral lymphatic vessel or peritumoral and intratumoral blood vessel area, and SLN-positive was found. CONCLUSIONS: Our results, taking into account the expression of either VEGF-C or related histopathological markers, indicated the possibility to use VEGF-C immunohistochemistry as a marker of metastatic progression, especially in thin cutaneous melanomas.


Subject(s)
Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Neoplastic , Melanoma , Receptors, CCR7/biosynthesis , Skin Neoplasms , Vascular Endothelial Growth Factor C/biosynthesis , Adult , Aged , Female , Humans , Lymphatic Metastasis , Male , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Retrospective Studies , Sentinel Lymph Node Biopsy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
8.
Polymers (Basel) ; 14(22)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36432947

ABSTRACT

Analysing the composition and organisation of the fibrous capsule formed as a result of the Foreign Body Response (FBR) to medical devices, is imperative for medical device improvement and biocompatibility. Typically, analysis is performed using histological techniques which often involve random sampling strategies. This method is excellent for acquiring representative values but can miss the unique spatial distribution of features in 3D, especially when analysing devices used in large animal studies. To overcome this limitation, we demonstrate a non-destructive method for high-resolution large sample imaging of the fibrous capsule surrounding human-sized implanted devices using diffusion tensor imaging (DTI). In this study we analyse the fibrous capsule surrounding two unique macroencapsulation devices that have been implanted in a porcine model for 21 days. DTI is used for 3D visualisation of the microstructural organisation and validated using the standard means of fibrous capsule investigation; histological analysis and qualitative micro computed tomography (microCT) and scanning electron microscopy (SEM) imaging. DTI demonstrated the ability to distinguish microstructural differences in the fibrous capsules surrounding two macroencapsulation devices made from different materials and with different surface topographies. DTI-derived metrics yielded insight into the microstructural organisation of both capsules which was corroborated by microCT, SEM and histology. The non-invasive characterisation of the integration of implants in the body has the potential to positively influence analysis methods in pre-clinical studies and accelerate the clinical translation of novel implantable devices.

9.
Adv Healthc Mater ; 10(14): e2100229, 2021 07.
Article in English | MEDLINE | ID: mdl-34165264

ABSTRACT

Medical devices, such as silicone-based prostheses designed for soft tissue implantation, often induce a suboptimal foreign-body response which results in a hardened avascular fibrotic capsule around the device, often leading to patient discomfort or implant failure. Here, it is proposed that additive manufacturing techniques can be used to deposit durable coatings with multiscale porosity on soft tissue implant surfaces to promote optimal tissue integration. Specifically, the "liquid rope coil effect", is exploited via direct ink writing, to create a controlled macro open-pore architecture, including over highly curved surfaces, while adapting atomizing spray deposition of a silicone ink to create a microporous texture. The potential to tailor the degree of tissue integration and vascularization using these fabrication techniques is demonstrated through subdermal and submuscular implantation studies in rodent and porcine models respectively, illustrating the implant coating's potential applications in both traditional soft tissue prosthetics and active drug-eluting devices.


Subject(s)
Prostheses and Implants , Silicones , Animals , Humans , Materials Testing , Porosity , Swine
10.
J Dermatol Sci ; 98(3): 186-194, 2020 06.
Article in English | MEDLINE | ID: mdl-32402513

ABSTRACT

BACKGROUND: Clinical skin manifestations are common in diabetes; however, molecular mechanisms underlying such defects are largely unknown. Several findings indicate a role for microRNAs (miRNAs) in skin homeostasis. OBJECTIVE: To investigate whether miRNA expression is altered in diabetic skin. METHODS: Type 1 and 2 mouse models of diabetes were used. MiRNA profiling was performed on RNA extracted from the skin of type 1 diabetic mice and non-diabetic controls. Expression levels of pri-miRNAs and of miRNA-biogenesis genes were also analyzed. Biogenesis gene expression analysis was performed in human dermal fibroblasts cultured in hyperglycemic, hypoxic or oxidative stress conditions. RESULTS: Several miRNAs were differentially expressed in diabetic skin with a general down-modulation as compared to controls. Bioinformatics analysis of signature-miRNA target genes showed the enrichment in pathways involved in skin homeostasis, such as TGF-ß and Wnt. MiRNA alteration in diabetic skin associated with reduced expression levels of DROSHA, DGCR8, XPO5, DICER1, AGO2, both as mRNA and protein. Reduced biogenesis gene expression did not correlate with accumulation of pri-miRNAs, which displayed differences in expression levels similar to those found for their mature miRNAs. Experiments with cultured fibroblasts showed that hypoxia and oxidative stress induced the down-regulation of miRNA-biogenesis genes in this skin cell type. CONCLUSION: A general down-regulation of differentially expressed miRNAs was found in diabetic skin. This alteration is part of and is dependent from a wider transcriptional defect also affecting the expression of pri-miRNAs and of genes responsible for miRNA biogenesis. Such an alteration is likely contributing to diabetic skin manifestations.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Hyperglycemia/complications , MicroRNAs/biosynthesis , Skin Diseases/pathology , Animals , Biopsy , Cell Hypoxia/genetics , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/chemically induced , Down-Regulation , Fibroblasts , Gene Expression Profiling , Gene Expression Regulation , Humans , Hyperglycemia/blood , Hyperglycemia/chemically induced , Hyperglycemia/genetics , Male , Mice , Oligonucleotide Array Sequence Analysis , Oxidative Stress/genetics , Signal Transduction/genetics , Skin/cytology , Skin/pathology , Skin Diseases/blood , Skin Diseases/etiology
11.
Adv Healthc Mater ; 9(11): e2000305, 2020 06.
Article in English | MEDLINE | ID: mdl-32339411

ABSTRACT

Regenerative medicine approaches, specifically stem cell technologies, have demonstrated significant potential to treat a diverse array of pathologies. However, such approaches have resulted in a modest clinical benefit, which may be attributed to poor cell retention/survival at the disease site. A delivery system that facilitates regional and repeated delivery to target tissues can provide enhanced clinical efficacy of cell therapies when localized delivery of high doses of cells is required. In this study, a new regenerative reservoir platform (Regenervoir) is described for use in large animal models, with relevance to cardiac, abdominal, and soft tissue pathologies. Regenervoir incorporates multiple novel design features essential for clinical translation, with a focus on scalability, mechanism of delivery, fixation to target tissue, and filling/refilling with a therapeutic cargo, and is demonstrated in an array of clinical applications that are easily translated to human studies. Regenervoir consists of a porous reservoir fabricated from a single material, a flexible thermoplastic polymer, capable of delivering cargo via fill lines to target tissues. A radiopaque shear thinning hydrogel can be delivered to the therapy reservoir and multiple fixation methods (laparoscopic tacks and cyanoacrylate bioadhesive) can be used to secure Regenervoir to target tissues through a minimally invasive approach.


Subject(s)
Hydrogels , Regenerative Medicine , Animals , Humans , Models, Animal , Polymers , Prostheses and Implants
12.
Matrix Biol ; 81: 3-16, 2019 08.
Article in English | MEDLINE | ID: mdl-30528862

ABSTRACT

Loss-of-function mutations in the gene encoding type VII collagen underlie recessive dystrophic epidermolysis bullosa (RDEB), a disease characterized by skin and mucosal blistering, impaired wound healing, and diffuse dermal inflammation and fibrosis. Transforming growth factor-ß signaling plays a crucial role in determining RDEB fibrotic microenvironment that leads to the development of disabling secondary disease manifestations, including hand and foot deformities. Experimental findings indicate that expression levels of decorin, a small leucine-rich proteoglycan and an endogenous TGF-ß inhibitor, can modulate RDEB disease phenotype by contrasting dermal fibroblast fibrotic behavior. In this study, the ability of decorin to modify RDEB course was investigated by systemically treating RDEB mice with a lentivirus expressing human decorin. Overexpressed decorin was able to enhance survival, and to limit digit contraction and the development of paw deformities. These effects were associated with decreased TGF-ß1 levels and TGF-ß signaling activation. Fibrotic traits were strongly reduced in paw skin and also attenuated in the non-chronically injured back skin. However, the expression of pro-inflammatory proteins was not decreased in both paw and back skin. Our findings confirm TGF-ß role in promoting fibrosis and disease progression in RDEB, and show that decorin counteracts disease manifestations by inhibiting TGF-ß activation. More generally, our data indicate that modifying extracellular matrix composition is an option to improve RDEB disease course.


Subject(s)
Decorin/genetics , Epidermolysis Bullosa Dystrophica/therapy , Genetic Vectors/administration & dosage , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Disease Progression , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Humans , Lentivirus/genetics , Mice , Signal Transduction , Survival Analysis , Transforming Growth Factor beta/metabolism , Treatment Outcome
13.
Biochimie ; 138: 106-115, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28479106

ABSTRACT

Interleukin-22 (IL-22) belongs to the family of IL-10 cytokines and is involved in a wide number of human diseases, including inflammatory disorders and cancer pathology. The ligand-receptor complex IL-22/IL-22R plays a key role in several pathways especially in the regulation and resolution of immune responses. The identification of novel compounds able to modulate IL-22/IL-22R complex could open the route to new therapeutic strategies in multiple human diseases. In this study, we designed and characterized IL-22 derived peptides at protein interface regions: several sequences revealed able to interfere with the protein complex with IC50 in the micromolar range as evaluated through Surface Plasmon Resonance (SPR) experiments. Their conformational characterization was carried out through Circular Dichroism (CD) and Nuclear Magnetic Resonance (NMR) spectroscopies, shedding new light into the features of IL-22 fragments and on structural determinants of IL-22/IL-22R1 recognition. Finally, several peptides were tested on human keratinocyte cultures for evaluating their ability to mimic the activation of molecular pathways downstream to IL-22R in response to IL-22 binding.


Subject(s)
Interleukins/chemistry , Models, Molecular , Amino Acid Sequence , Humans , Interleukins/metabolism , Keratinocytes/metabolism , Magnetic Resonance Spectroscopy , Peptides , Protein Structure, Tertiary , Signal Transduction , Surface Plasmon Resonance , Interleukin-22
14.
J Invest Dermatol ; 126(2): 315-24, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16374478

ABSTRACT

The multidomain serine protease inhibitor lymphoepithelial Kazal-type related inhibitor (LEKTI) represents a key regulator of the proteolytic events occurring during epidermal barrier formation and hair development, as attested by the severe autosomal recessive ichthyosiform skin condition Netherton syndrome (NS) caused by mutations in its encoding gene, serine protease inhibitor Kazal-type 5 (SPINK5). Synthesized as a proprotein, LEKTI is rapidly cleaved intracellularly, thus generating a number of potentially bioactive fragments that are secreted. Here, we show that SPINK5 generates three classes of transcripts encoding three different LEKTI isoforms, which differ in their C-terminal portion. In addition to the previously described 15 domain isoform, SPINK5 encodes a shorter LEKTI isoform composed of only the first 13 domains, as well as a longer isoform carrying a 30-amino-acid residue insertion between the 13th and 14th inhibitory domains. We demonstrate that variable amounts of SPINK5 alternative transcripts are detected in all SPINK5 transcriptionally active tissues. Finally, we show that in differentiated cultured human keratinocytes all SPINK5 alternative transcripts are translated into protein and that the LEKTI precursors generate distinct secreted C-terminal proteolytic fragments from a similar cleavage site. Since several data indicate a biological role for the pro-LEKTI-cleaved polypeptides, we hypothesize that the alternative processing of the SPINK5 pre-messenger RNA represents an additional mechanism to further increase the structural and functional diversity of the LEKTI bioactive fragments.


Subject(s)
Carrier Proteins/genetics , Ichthyosis/genetics , RNA Precursors/metabolism , Amino Acid Sequence , Cell Differentiation , Cells, Cultured , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Molecular Sequence Data , Protein Biosynthesis/genetics , Protein Isoforms/genetics , Proteinase Inhibitory Proteins, Secretory , RNA, Messenger/genetics , Serine Peptidase Inhibitor Kazal-Type 5 , Syndrome , Transcription, Genetic
15.
J Dermatol Sci ; 41(1): 11-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16216466

ABSTRACT

The placenta growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family that has been shown to play an important role in promoting adult pathological angiogenesis. Besides inducing its own signaling in endothelial cells, PlGF exerts its angiogenic action by synergising with VEGF. In the skin, PlGF expression is upregulated during wound healing and PlGF-deficient mice show delayed wound closure, indicating that this factor promotes angiogenesis during skin repair. Moreover, PlGF expression by melanoma cells has been linked to tumor growth. The analysis of a transgenic mouse model constitutively expressing high levels of PlGF in basal keratinocytes has shown that this factor has strong angiogenic properties in the skin during both embryonic and post-natal life. Furthermore, PlGF delivery to the skin via an adenoviral vector induces the formation of large and stable blood vessels, but contrary to VEGF application, does not affect lymphatic vessel functionality. Such evidence opens the possibility of employing PlGF for therapeutic modulation of skin angiogenesis.


Subject(s)
Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Pregnancy Proteins/physiology , Skin/blood supply , Animals , Humans , Neovascularization, Physiologic/drug effects , Placenta Growth Factor , Pregnancy Proteins/pharmacology
16.
J Invest Dermatol ; 135(11): 2862-2870, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26168231

ABSTRACT

Impaired re-epithelialization, imbalanced expression of cytokines and growth factors, and vascular disease contribute to healing impairment in diabetes. IL-22, a pro-inflammatory cytokine mediating a cross-talk between immune system and epithelial cells, has been shown to have a role in repair processes. In this study we aimed to investigate IL-22 regenerative potential in the poor healing context of diabetic wounds. By using streptozotocin-induced diabetic mice, we demonstrated that IL-22 wound treatment significantly accelerated the healing process, by promoting re-epithelialization, granulation tissue formation, and vascularization. Improved re-epithelialization was associated with increased keratinocyte proliferation and signal transducer and activator of transcription 3 (STAT3) activation. We showed that endogenous IL-22 content was reduced at both mRNA and protein level during the inflammatory phase of diabetic wounds, with fewer IL-22-positive cells infiltrating the granulation tissue. We demonstrated that IL-22 treatment promoted proliferation and injury repair of hyperglycemic keratinocytes and induced activation of STAT3 and extracellular signal-regulated kinase transduction pathways in keratinocytes grown in hyperglycemic condition or isolated from diabetic patients. Finally, we demonstrated that IL-22 treatment was able to inhibit diabetic keratinocyte differentiation while promoting vascular endothelial growth factor release. Our data indicate a pro-healing role of IL-22 in diabetic wounds, suggesting a therapeutic potential for this cytokine in diabetic ulcer management.


Subject(s)
Interleukins/pharmacology , Keratinocytes/drug effects , Skin Ulcer/drug therapy , Wound Healing/physiology , Administration, Topical , Animals , Biopsy, Needle , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Interleukins/metabolism , Keratinocytes/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , Random Allocation , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Skin Ulcer/metabolism , Skin Ulcer/pathology , Vascular Endothelial Growth Factor A/metabolism , Interleukin-22
17.
FEBS J ; 282(23): 4435-49, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26361888

ABSTRACT

The HECT-type E3 ubiquitin ligase Itch is absent in the non-agouti-lethal 18H or Itchy mice, which develop a severe immunological disease. Several of the known Itch substrates are relevant for epidermal development and homeostasis, such as p63, Notch, c-Jun and JunB. By analysing Itchy mice before the onset of immunological alterations, we investigated the contribution of Itch in skin development and wound healing. Itchy newborn mice manifested hyperplastic epidermis, which is not present in adulthood. Itch(-/-) cultured keratinocytes showed overexpression of proliferating markers and increased capability to proliferate, migrate and to repair a scratch injury in vitro. These data correlated with improved in vivo wound healing in Itchy mice, at late time points of the repair process when Itch is physiologically upregulated. Despite healing acceleration, epidermal remodelling was delayed in the scars of Itch(-/-) mice, as indicated by enhanced epidermal thickening, keratinocyte proliferation and keratin 6 expression, and retarded keratin 14 polarization to the basal layer. Itch(-/-) keratinocyte prolonged activation was not associated with increased immune cell persistence in the scars. Our in vitro and in vivo results indicate that Itch plays a role in epidermal homeostasis and remodelling and this feature does not seem to depend on immunological alterations.


Subject(s)
Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism , Wound Healing , Animals , Cell Proliferation , Cells, Cultured , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Knockout
18.
J Invest Dermatol ; 131(9): 1821-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21654836

ABSTRACT

The 14-3-3 protein family controls diverse biochemical processes through interaction with phosphorylated consensus sequences in protein targets. Its epithelial specific member, 14-3-3σ, also known as stratifin, is highly expressed in differentiated keratinocytes, and in vitro evidence indicates that 14-3-3σ downregulation leads to keratinocyte immortalization. To define the role of 14-3-3σ in skin homeostasis in vivo, we generated transgenic mice overexpressing 14-3-3σ in proliferating keratinocytes of the epidermis and hair follicle. Transgenic animals show decreased epidermal thickness and hair follicle density associated with reduced number of proliferating keratinocytes and decreased levels of keratins 14, 5, and 15. Primary keratinocytes isolated from transgenic mice manifest reduced proliferation and migration. Moreover, clonogenicity assessment and label-retaining analysis reveal a reduction in keratinocyte progenitor cell number in transgenic mice. Response to IGF-1 is strongly impaired in cultured transgenic keratinocytes compared with wild-type cells. Consistently, activation of phosphoinositol 3-kinase (PI3K), AKT, and Rac1, all IGF-1 downstream mediators, is reduced. Our results demonstrate that 14-3-3σ controls the in vivo epidermal proliferation-differentiation switch by reducing proliferative potential and forcing keratinocytes to exit the cell cycle, and that this effect associates with inhibition of the IGF-1 pathway.


Subject(s)
14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Epidermis/physiology , Exonucleases/genetics , Exonucleases/metabolism , Hair Follicle/physiology , Keratinocytes/physiology , Animals , Cell Division/physiology , Cells, Cultured , Clone Cells/cytology , Clone Cells/physiology , Epidermal Cells , Exoribonucleases , Hair Follicle/cytology , Insulin-Like Growth Factor I/metabolism , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Phenotype , Promoter Regions, Genetic/physiology , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology
19.
J Invest Dermatol ; 130(1): 93-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19641516

ABSTRACT

Experimental evidence suggests that in autoimmune thyroid diseases (AITDs) the skin is a target of autoantibodies against thyroid-specific antigens; however, the role of these autoantibodies in skin alterations remains unclear. To gain insight into the function of nominally thyroid-specific genes in skin, we analyzed the expression of thyroid-stimulating hormone-receptor (TSH-R), thyroglobulin (Tg), sodium iodide symporter (NIS), and thyroperoxidase (TPO) genes in normal human skin biopsies and cultured primary keratinocytes and dermal fibroblasts. The results revealed the presence of all the transcripts in skin biopsies. However, in keratinocytes and fibroblasts, only TSH-R messenger RNA was always detected. Western blot and immunohistochemical analyses of skin specimens confirmed the presence of TSH-R protein in keratinocytes and fibroblasts. Moreover, TSH treatment induced the proliferation of cultured keratinocytes and fibroblasts and increased keratinocyte intracellular cAMP. Finally, affinity-purified IgGs from serum of patients affected by Graves' disease, but not by chronic lymphocytic thyroiditis, stimulated cAMP accumulation in cultured keratinocytes, as well as their proliferation. In conclusion, the expression of thyroid-specific genes in cultured keratinocytes and fibroblasts and the mitogenic effects of TSH and IgGs on these cells support the concept that autoantibodies against thyroid-specific antigens may contribute to cutaneous symptoms in AITDs.


Subject(s)
Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Skin/cytology , Skin/immunology , Thyroid Diseases , Autoantibodies/blood , Autoantigens/genetics , Autoantigens/immunology , Autoantigens/metabolism , Biopsy , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression/physiology , Humans , Immunoglobulin G/blood , Iodide Peroxidase/genetics , Iodide Peroxidase/immunology , Iodide Peroxidase/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/immunology , Iron-Binding Proteins/metabolism , Keratinocytes/cytology , Keratinocytes/physiology , RNA, Messenger/metabolism , Receptors, Thyrotropin/immunology , Skin/metabolism , Symporters/genetics , Symporters/immunology , Symporters/metabolism , Thyroglobulin/genetics , Thyroglobulin/immunology , Thyroglobulin/metabolism , Thyroid Diseases/immunology , Thyroid Diseases/metabolism , Thyroid Diseases/physiopathology , Thyrotropin/genetics , Thyrotropin/immunology , Thyrotropin/metabolism
20.
J Clin Invest ; 119(12): 3573-85, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19920355

ABSTRACT

Th subsets are defined according to their production of lineage-indicating cytokines and functions. In this study, we have identified a subset of human Th cells that infiltrates the epidermis in individuals with inflammatory skin disorders and is characterized by the secretion of IL-22 and TNF-alpha, but not IFN-gamma, IL-4, or IL-17. In analogy to the Th17 subset, cells with this cytokine profile have been named the Th22 subset. Th22 clones derived from patients with psoriasis were stable in culture and exhibited a transcriptome profile clearly separate from those of Th1, Th2, and Th17 cells; it included genes encoding proteins involved in tissue remodeling, such as FGFs, and chemokines involved in angiogenesis and fibrosis. Primary human keratinocytes exposed to Th22 supernatants expressed a transcriptome response profile that included genes involved in innate immune pathways and the induction and modulation of adaptive immunity. These proinflammatory Th22 responses were synergistically dependent on IL-22 and TNF-alpha. Furthermore, Th22 supernatants enhanced wound healing in an in vitro injury model, which was exclusively dependent on IL-22. In conclusion, the human Th22 subset may represent a separate T cell subset with a distinct identity with respect to gene expression and function, present within the epidermal layer in inflammatory skin diseases. Future strategies directed against the Th22 subset may be of value in chronic inflammatory skin disorders.


Subject(s)
Epidermal Cells , Epidermis/immunology , Interleukins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Clone Cells , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Gene Expression Profiling , Humans , Immunity, Innate , In Vitro Techniques , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Interleukins/genetics , Keratinocytes/immunology , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Receptors, CCR10/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , Tumor Necrosis Factor-alpha/metabolism , Wound Healing/genetics , Wound Healing/immunology , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL