Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(9): 1034-1045, 2020 09.
Article in English | MEDLINE | ID: mdl-32661363

ABSTRACT

Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Macrophages/immunology , Microbiota/immunology , Neutrophils/immunology , Skin/immunology , Animals , Cells, Cultured , Chemokine CXCL10/metabolism , Humans , Immunity, Innate , Inflammation , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology , Symbiosis , Wound Healing
2.
J Cell Sci ; 135(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35293576

ABSTRACT

Vertebrate lonesome kinase (VLK) is the only known extracellular tyrosine kinase, but its physiological functions are largely unknown. We show that VLK is highly expressed in hepatocytes of neonatal mice, but downregulated during adulthood. To determine the role of VLK in liver homeostasis and regeneration, we generated mice with a hepatocyte-specific knockout of the VLK gene (Pkdcc). Cultured progenitor cells established from primary hepatocytes of Pkdcc knockout mice produced a secretome, which promoted their own proliferation in 3D spheroids and proliferation of cultured fibroblasts. In vivo, Pkdcc knockout mice developed liver steatosis with signs of inflammation and perivascular fibrosis upon aging, combined with expansion of liver progenitor cells. In response to chronic CCl4-induced liver injury, the pattern of deposited collagen was significantly altered in these mice. The liver injury marker alpha-fetoprotein (AFP) was increased in the secretome of VLK-deficient cultured progenitor cells and in liver tissues of aged or CCl4-treated knockout mice. These results support a key role for VLK and extracellular protein phosphorylation in liver homeostasis and repair through paracrine control of liver cell function and regulation of appropriate collagen deposition. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Hepatocytes , Secretome , Adult , Aged , Animals , Collagen/metabolism , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Mice, Knockout , Protein-Tyrosine Kinases/metabolism
3.
Cell Mol Life Sci ; 80(4): 89, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36920550

ABSTRACT

Vertebrate lonesome kinase (VLK) is the only known secreted tyrosine kinase and responsible for the phosphorylation of a broad range of secretory pathway-resident and extracellular matrix proteins. However, its cell-type specific functions in vivo are still largely unknown. Therefore, we generated mice lacking the VLK gene (protein kinase domain containing, cytoplasmic (Pkdcc)) in mesenchymal cells. Most of the homozygous mice died shortly after birth, most likely as a consequence of their lung abnormalities and consequent respiratory failure. E18.5 embryonic lungs showed a reduction of alveolar type II cells, smaller bronchi, and an increased lung tissue density. Global mass spectrometry-based quantitative proteomics identified 97 proteins with significantly and at least 1.5-fold differential abundance between genotypes. Twenty-five of these had been assigned to the extracellular region and 15 to the mouse matrisome. Specifically, fibromodulin and matrilin-4, which are involved in extracellular matrix organization, were significantly more abundant in lungs from Pkdcc knockout embryos. These results support a role for mesenchyme-derived VLK in lung development through regulation of matrix dynamics and the resulting modulation of alveolar epithelial cell differentiation.


Subject(s)
Extracellular Matrix , Protein Kinases , Animals , Mice , Protein Kinases/genetics , Organogenesis/genetics , Lung , Mesoderm , Vertebrates , Protein-Tyrosine Kinases
4.
Biochem Soc Trans ; 51(1): 101-111, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36762597

ABSTRACT

The transcription factor NRF2 is well known as a master regulator of the cellular stress response. As such, activation of NRF2 has gained widespread attention for its potential to prevent tissue injury, but also as a possible therapeutic approach to promote repair processes. While NRF2 activation affects most or even all cell types, its effect on epithelial cells during repair processes has been particularly well studied. In response to tissue injury, these cells proliferate, migrate and/or spread to effectively repair the damage. In this review, we discuss how NRF2 governs repair of epithelial tissues, and we highlight the increasing number of NRF2 targets with diverse roles in regulating epithelial repair.


Subject(s)
NF-E2-Related Factor 2 , Signal Transduction , NF-E2-Related Factor 2/metabolism , Signal Transduction/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Gene Expression Regulation , Epithelial Cells/metabolism , Oxidative Stress
5.
Nat Chem Biol ; 17(8): 865-871, 2021 08.
Article in English | MEDLINE | ID: mdl-34253910

ABSTRACT

Collagens are fibrous proteins that are integral to the strength and stability of connective tissues. During collagen maturation, lysyl oxidases (LOX) initiate the cross-linking of fibers, but abnormal LOX activity is associated with impaired tissue function as seen in fibrotic and malignant diseases. Visualizing and targeting this dynamic process in healthy and diseased tissue is important, but so far not feasible. Here we present a probe for the simultaneous monitoring and targeting of LOX-mediated collagen cross-linking that combines a LOX-activity sensor with a collagen peptide to chemoselectively target endogenous aldehydes generated by LOX. This synergistic probe becomes covalently anchored and lights up in vivo and in situ in response to LOX at the sites where cross-linking occurs, as demonstrated by staining of normal skin and cancer sections. We anticipate that our reactive collagen-based sensor will improve understanding of collagen remodeling and provide opportunities for the diagnosis of fibrotic and malignant diseases.


Subject(s)
Collagen/metabolism , Cross-Linking Reagents/metabolism , Peptides/metabolism , Protein-Lysine 6-Oxidase/metabolism , Aldehydes/chemistry , Aldehydes/metabolism , Animals , Collagen/chemistry , Cross-Linking Reagents/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Peptides/chemistry , Protein-Lysine 6-Oxidase/chemistry
6.
Cell ; 132(5): 818-31, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18329368

ABSTRACT

Mammalian cells export most proteins by the endoplasmic reticulum/Golgi-dependent pathway. However, some proteins are secreted via unconventional, poorly understood mechanisms. The latter include the proinflammatory cytokines interleukin(IL)-1beta, IL-18, and IL-33, which require activation by caspase-1 for biological activity. Caspase-1 itself is activated by innate immune complexes, the inflammasomes. Here we show that secretion of the leaderless proteins proIL-1alpha, caspase-1, and fibroblast growth factor (FGF)-2 depends on caspase-1 activity. Although proIL-1alpha and FGF-2 are not substrates of the protease, we demonstrated their physical interaction. Secretome analysis using iTRAQ proteomics revealed caspase-1-mediated secretion of other leaderless proteins with known or unknown extracellular functions. Strikingly, many of these proteins are involved in inflammation, cytoprotection, or tissue repair. These results provide evidence for an important role of caspase-1 in unconventional protein secretion. By this mechanism, stress-induced activation of caspase-1 directly links inflammation to cytoprotection, cell survival, and regenerative processes.


Subject(s)
Caspase 1/immunology , Caspase 1/metabolism , Animals , COS Cells , Caspase 1/genetics , Cell Line , Chlorocebus aethiops , Fibroblast Growth Factor 2/metabolism , Humans , Inflammation/immunology , Interleukin-1alpha/metabolism , Keratinocytes/immunology , Macrophages/immunology , Mice , Protein Binding , Protein Sorting Signals , Proteomics , RNA, Small Interfering/genetics
7.
Nucleic Acids Res ; 49(7): 3748-3763, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33764436

ABSTRACT

Epigenetic regulation of cell and tissue function requires the coordinated action of transcription factors. However, their combinatorial activities during regeneration remain largely unexplored. Here, we discover an unexpected interaction between the cytoprotective transcription factor NRF2 and p63- a key player in epithelial morphogenesis. Chromatin immunoprecipitation combined with sequencing and reporter assays identifies enhancers and promoters that are simultaneously activated by NRF2 and p63 in human keratinocytes. Modeling of p63 and NRF2 binding to nucleosomal DNA suggests their chromatin-assisted interaction. Pharmacological and genetic activation of NRF2 increases NRF2-p63 binding to enhancers and promotes keratinocyte proliferation, which involves the common NRF2-p63 target cyclin-dependent kinase 12. These results unravel a collaborative function of NRF2 and p63 in the control of epidermal renewal and suggest their combined activation as a strategy to promote repair of human skin and other stratified epithelia.


Subject(s)
Keratinocytes , NF-E2-Related Factor 2/physiology , Skin , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinases/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Skin/cytology , Skin/metabolism
8.
Eur J Immunol ; 50(9): 1335-1349, 2020 09.
Article in English | MEDLINE | ID: mdl-32306381

ABSTRACT

Wound healing involves the concerted action of various lymphoid and in particular myeloid cell populations. To characterize and quantitate different types of myeloid cells and to obtain information on their kinetics during wound healing, we performed multiparametric flow cytometry analysis. In healthy mice, neutrophil numbers increased early after injury and returned to near basal levels after completion of healing. Macrophages, monocyte-derived dendritic cells (DCs), and eosinophils were abundant throughout the healing phase, in particular in early wounds, and Langerhans cells increased after wounding and remained elevated after epithelial closure. Major differences in healing-impaired diabetic mice were a much higher percentage of immune cells in late wounds, mainly as a result of neutrophil, macrophage, and monocyte persistence; reduced numbers and percentages of macrophages and monocyte-derived DCs in early wounds; and of Langerhans cells, conventional DCs, and eosinophils throughout the healing process. Finally, unbiased cluster analysis (PhenoGraph) identified a large number of different clusters of myeloid cells in skin wounds. These results provide insight into myeloid cell diversity and dynamics during wound repair and highlight the abnormal inflammatory response associated with impaired healing.


Subject(s)
Myeloid Cells/physiology , Wound Healing/physiology , Animals , Diabetes Complications/immunology , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental , Mice, Inbred C57BL
9.
J Cell Mol Med ; 24(2): 1774-1785, 2020 01.
Article in English | MEDLINE | ID: mdl-31830366

ABSTRACT

Fibroblast growth factors (FGFs) are key regulators of tissue development, homeostasis and repair, and abnormal FGF signalling is associated with various human diseases. In human and murine epidermis, FGF receptor 3 (FGFR3) activation causes benign skin tumours, but the consequences of FGFR3 deficiency in this tissue have not been determined. Here, we show that FGFR3 in keratinocytes is dispensable for mouse skin development, homeostasis and wound repair. However, the defect in the epidermal barrier and the resulting inflammatory skin disease that develops in mice lacking FGFR1 and FGFR2 in keratinocytes were further aggravated upon additional loss of FGFR3. This caused fibroblast activation and fibrosis in the FGFR1/FGFR2 double-knockout mice and even more in mice lacking all three FGFRs, revealing functional redundancy of FGFR3 with FGFR1 and FGFR2 for maintaining the epidermal barrier. Taken together, our study demonstrates that FGFR1, FGFR2 and FGFR3 act together to maintain epidermal integrity and cutaneous homeostasis, with FGFR2 being the dominant receptor.


Subject(s)
Keratinocytes/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cells, Cultured , Epidermis/metabolism , Female , Fibrosis , Homeostasis , Keratinocytes/pathology , Mice, Knockout , Phenotype , Signal Transduction , Wound Healing
10.
Development ; 144(22): 4047-4060, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29138288

ABSTRACT

Tissue injury initiates a complex repair process, which in some organisms can lead to the complete regeneration of a tissue. In mammals, however, the repair of most organs is imperfect and results in scar formation. Both regeneration and repair are orchestrated by a highly coordinated interplay of different growth factors and cytokines. Among the key players are the fibroblast growth factors (FGFs), which control the migration, proliferation, differentiation and survival of different cell types. In addition, FGFs influence the expression of other factors involved in the regenerative response. Here, we summarize current knowledge on the roles of endogenous FGFs in regeneration and repair in different organisms and in different tissues and organs. Gaining a better understanding of these FGF activities is important for appropriate modulation of FGF signaling after injury to prevent impaired healing and to promote organ regeneration in humans.


Subject(s)
Fibroblast Growth Factors/metabolism , Regeneration/physiology , Wound Healing , Animals , Humans , Models, Biological , Organ Specificity , Signal Transduction
11.
Am J Pathol ; 189(3): 568-579, 2019 03.
Article in English | MEDLINE | ID: mdl-30593821

ABSTRACT

The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor is a key regulator of the cellular stress response. Therefore, pharmacologic Nrf2 activation is a promising strategy for skin protection and cancer prevention. This study found that genetic Nrf2 activation in keratinocytes accelerates wound repair. Enhanced proliferation of cells of the pilosebaceous unit peripheral to the wound and a concomitant acceleration of re-epithelialization were identified as the underlying mechanism. Nrf2 specifically promoted the expansion of pilosebaceous cells expressing markers of junctional zone and upper isthmus follicular stem cells. This may result, at least in part, from the up-regulation of the direct Nrf2 target epigen and a concomitant increase in epidermal growth factor receptor signaling. The increase in pilosebaceous cells provided a larger pool of keratinocytes that migrate into the wound, resulting in faster wound closure. These results unravel a novel function of Nrf2 in wound repair and suggest the use of NRF2-activating compounds in patients with impaired healing.


Subject(s)
Gene Expression Regulation , Keratinocytes/metabolism , NF-E2-Related Factor 2/metabolism , Re-Epithelialization , Signal Transduction , Skin/metabolism , Animals , Keratinocytes/pathology , Mice , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Skin/pathology
12.
Nat Rev Mol Cell Biol ; 9(8): 628-38, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18628784

ABSTRACT

What is the relationship between the wound-healing process and the development of cancer? Malignant tumours often develop at sites of chronic injury, and tissue injury has an important role in the pathogenesis of malignant disease, with chronic inflammation being the most important risk factor. The development and functional characterization of genetically modified mice that lack or overexpress genes that are involved in repair, combined with gene-expression analysis in wounds and tumours, have highlighted remarkable similarities between wound repair and cancer. However, a few crucial differences were also observed, which could account for the altered metabolism, impaired differentiation capacity and invasive growth of malignant tumours.


Subject(s)
Models, Biological , Neoplasms/etiology , Wound Healing/physiology , Animals , Cicatrix/pathology , Epithelium/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Fibrin/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Inflammation/pathology , Neoplasms/genetics , Neovascularization, Physiologic/physiology , Wound Healing/genetics
13.
Eur J Immunol ; 48(6): 1001-1013, 2018 06.
Article in English | MEDLINE | ID: mdl-29457218

ABSTRACT

Healing of skin wounds is orchestrated by various types of immune cells, but little is known about the role of FoxP3+ regulatory T cells (Tregs) in this process. Here, we determined if Tregs are important for wound healing in normal mice and if they contribute to the accelerated healing of mice overexpressing the growth and differentiation factor activin. Diphtheria toxin induced Treg depletion prior to injury caused impaired healing characterized by delayed reepithelialization, reduced wound contraction, and impaired vessel maturation. The accelerated wound repair of activin-transgenic mice was also abrogated. Mechanistically, we found a strong increase in IL-4 levels combined with overrepresentation of T-bet+ and GATA-3+ αß T cells in Treg-depleted 7-day wounds. In addition, numbers of IFN-γ- or IL-17A-producing CD4+ and CD4- T cells were elevated. These results demonstrate that Treg depletion in wounds facilitates the expansion of an αß T-cell population with features of Th1 and Th2 cells, and suggest that concomitant changes in the cytokine milieu disturb the healing process.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Wound Healing/immunology , Activins/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Diphtheria Toxin/immunology , Forkhead Transcription Factors/genetics , GATA3 Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , T-Box Domain Proteins/genetics , Wound Healing/genetics
14.
PLoS Genet ; 12(1): e1005800, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26808544

ABSTRACT

The tripeptide glutathione is the most abundant cellular antioxidant with high medical relevance, and it is also required as a co-factor for various enzymes involved in the detoxification of reactive oxygen species and toxic compounds. However, its cell-type specific functions and its interaction with other cytoprotective molecules are largely unknown. Using a combination of mouse genetics, functional cell biology and pharmacology, we unraveled the function of glutathione in keratinocytes and its cross-talk with other antioxidant defense systems. Mice with keratinocyte-specific deficiency in glutamate cysteine ligase, which catalyzes the rate-limiting step in glutathione biosynthesis, showed a strong reduction in keratinocyte viability in vitro and in the skin in vivo. The cells died predominantly by apoptosis, but also showed features of ferroptosis and necroptosis. The increased cell death was associated with increased levels of reactive oxygen and nitrogen species, which caused DNA and mitochondrial damage. However, epidermal architecture, and even healing of excisional skin wounds were only mildly affected in the mutant mice. The cytoprotective transcription factor Nrf2 was strongly activated in glutathione-deficient keratinocytes, but additional loss of Nrf2 did not aggravate the phenotype, demonstrating that the cytoprotective effect of Nrf2 is glutathione dependent. However, we show that deficiency in glutathione biosynthesis is efficiently compensated in keratinocytes by the cysteine/cystine and thioredoxin systems. Therefore, our study highlights a remarkable antioxidant capacity of the epidermis that ensures skin integrity and efficient wound healing.


Subject(s)
Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Skin/metabolism , Thioredoxins/genetics , Animals , Antioxidants/metabolism , Apoptosis/genetics , Cell Survival , DNA Damage/genetics , Glutamate-Cysteine Ligase/deficiency , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/biosynthesis , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , NF-E2-Related Factor 2/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism , Skin/pathology , Thioredoxins/metabolism , Wound Healing/genetics
15.
Int J Mol Sci ; 20(16)2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31398789

ABSTRACT

The nuclear factor-erythroid 2-related factor 2 (NRF2) transcription factor plays a central role in mediating the cellular stress response. Due to their antioxidant properties, compounds activating NRF2 have received much attention as potential medications for disease prevention, or even for therapy. Accumulating evidence suggests that activation of the NRF2 pathway also has a major impact on wound healing and may be beneficial in the treatment of chronic wounds, which remain a considerable health and economic burden. While NRF2 activation indeed shows promise, important considerations need to be made in light of corresponding evidence that also points towards pro-tumorigenic effects of NRF2. In this review, we discuss the evidence to date, highlighting recent advances using gain- and loss-of-function animal models and how these data fit with observations in humans.


Subject(s)
Gene Expression Regulation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Wound Healing , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytoprotection , Fibroblasts , Humans , Keratinocytes/metabolism , Loss of Function Mutation , Oxidative Stress , Phenotype
16.
Eur J Immunol ; 47(5): 806-817, 2017 05.
Article in English | MEDLINE | ID: mdl-28247911

ABSTRACT

The transcription factor Nrf2 regulates the expression of genes required for protection from xenobiotic and oxidative stress. Under normal conditions Nrf2 is constantly degraded upon ubiquitination, mediated by the Nrf2 inhibitor Keap1. Inflammasomes represent stress-induced protein complexes. They are critically involved in acute and chronic inflammation through caspase-1-mediated activation of pro-inflammatory cytokines. Here, we demonstrate that Nrf2 is a positive regulator of the NLRP3 inflammasome. In contrast, Nrf2-activating compounds, including the anti-inflammatory drug dimethyl fumarate (DMF), inhibit inflammasome activation. Both effects are independent of the transcriptional activity of Nrf2 and, at least in part, not interdependent. On the other hand, NLRP3 inflammasome activation induces a rapid and partly caspase-1- and Keap1-independent degradation of Nrf2. These data argue against a simultaneous activation of both stress-related pathways. Finally, we provide evidence that the cross-regulation of both pathways is controlled by a physical interaction between the Nrf2/Keap1 and NLRP3 complexes.


Subject(s)
Inflammasomes/immunology , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Caspase 1/metabolism , Cytokines/immunology , Dimethyl Fumarate/pharmacology , Gene Expression Regulation , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammation , Keratinocytes , Mice , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Signal Transduction/drug effects
18.
J Immunol ; 196(11): 4663-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183581

ABSTRACT

The Nrf2 transcription factor is well known for its cytoprotective functions through regulation of genes involved in the detoxification of reactive oxygen species or toxic compounds. Therefore, activation of Nrf2 is a promising strategy for the protection of tissues from various types of insults and for cancer prevention. However, recent studies revealed a proinflammatory activity of activated Nrf2 and a stimulating effect on epithelial cell proliferation, but the underlying mechanisms of action and the responsible target genes are largely unknown. Using a combination of gene expression profiling, chromatin immunoprecipitation, and targeted proteomics via selected reaction monitoring, we show that the gene encoding the proinflammatory cytokine IL-36γ is a novel direct target of Nrf2 in keratinocytes and hepatocytes in vitro and in vivo. As a consequence, upregulation of IL-36γ expression occurred upon genetic or pharmacological activation of Nrf2 in the epidermis and in the normal and regenerating liver. Functional in vitro studies demonstrate that IL-36γ directly stimulates proliferation of keratinocytes. In particular, it induces expression of keratinocyte mitogens in fibroblasts, suggesting that the Nrf2-IL-36γ axis promotes keratinocyte proliferation through a double paracrine loop. These results provide mechanistic insight into Nrf2 action in the control of inflammation and cell proliferation through regulation of a proinflammatory cytokine with a key function in various inflammatory diseases.


Subject(s)
Autocrine Communication , Cell Proliferation , Interleukin-1/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , NF-E2-Related Factor 2/metabolism , Paracrine Communication , Animals , Cells, Cultured , Interleukin-1/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic
19.
Semin Immunol ; 26(4): 321-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24556599

ABSTRACT

The attraction and activation of immune cells is an important response of the skin to injury and allows an efficient defense against invading pathogens. In addition, immune cells fulfill various functions that are important for the repair process. An exaggerated inflammatory response, however, is a hallmark of chronic, non-healing wounds. Therefore, it is essential to strictly control and coordinate the levels and activities of various immune cells in normal and wounded skin. Recent studies provided insight into the molecular mechanisms underlying the inflammatory response after wounding, and various transcriptional regulators involved in this process have been identified. This review summarizes our current knowledge on the function of different transcription factors in wound repair, with particular emphasis on proteins with a documented role in the control of wound inflammation.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Transcription, Genetic , Wound Healing , Animals , Blood Coagulation , Inflammation/immunology , Mice , Skin/injuries
20.
Genes Dev ; 24(10): 1045-58, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20478997

ABSTRACT

Ultraviolet (UV) B irradiation can severely damage the skin and even induce tumorigenesis. It exerts its effects by direct DNA modification and by formation of reactive oxygen species (ROS). We developed a strategy to genetically activate target gene expression of the transcription factor NF-E2-related factor 2 (Nrf2) in keratinocytes in vivo based on expression of a constitutively active Nrf2 mutant. Activation of Nrf2 target genes strongly reduced UVB cytotoxicity through enhancement of ROS detoxification. Remarkably, the protective effect was extended to neighboring cells. Using different combinations of genetically modified mice, we demonstrate that Nrf2 activates the production, recycling, and release of glutathione and cysteine by suprabasal keratinocytes, resulting in protection of basal cells in a paracrine, glutathione/cysteine-dependent manner. Most importantly, we found that endogenous Nrf2 controls selective protection of suprabasal keratinocytes from UVB-induced apoptosis through activation of cytoprotective genes. This finding explains the preferential UVB-induced apoptosis of basal cells, which is important for elimination of mutated stem cells as well as for preservation of skin integrity. Taken together, our results identify Nrf2 as a key regulator in the UV response of the skin.


Subject(s)
Cytoprotection/physiology , Glutathione/metabolism , Keratinocytes/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Female , Gene Expression Regulation , Humans , Keratinocytes/radiation effects , Mice , Mice, Transgenic , Mutation/genetics , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL