Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38407798

ABSTRACT

Due to their full differentiation capacity in vitro, the culture of human primary keratinocytes (HPKs) represents a physiological model for answering basic biological and dermatological research questions, including those related to skin diseases and the investigation of treatment options. When modified with the CRISPR/Cas9 gene editing approach and cultivated in organotypic 3D epidermal equivalents (EEs), these human cells have the potential to replace established mouse models. However, even when cultivated on feeder cells, HPKs have only a low proliferation capacity in 2D culture, limiting their application potential. This is particularly true for CRISPR/Cas9-modified HPKs, whose generation commonly requires selection of targeted cells, negatively affecting their lifespan. Here, we describe a robust protocol for the rapid, simple, and efficient generation of single- and multi-gene CRISPR/Cas9 knockout HPKs by electroporation of ribonucleoprotein (RNP) complexes, which comprise one or multiple guide RNAs (gRNAs) and Cas9 protein. Unlike DNA transfection or virus-based targeting strategies, electroporation of RNPs represents a targeting approach that minimizes immunological and toxic side effects. Using efficient gRNAs results in the generation of HPKs with a high yield of knockout cells, allowing for their immediate use in experiments without requiring the laborious process of selecting targeted cells or maintaining a feeder cell culture. Furthermore, the use of RNPs and their delivery via electroporation minimizes off-target and other unspecific effects, preventing unintended genomic alterations. Most importantly, CRISPR/Cas9 knockout HPKs generated with this protocol have the ability to form a fully differentiated epidermis in 3D, thus facilitating the understanding of specific protein functions in a highly physiological human skin model. Alternatively, this approach proves valuable for generating models of mono- or polygenic skin diseases via knockouts, providing insights into the underlying molecular mechanisms and facilitating the development of novel therapeutic approaches.

2.
J Invest Dermatol ; 142(9): 2313-2322, 2022 09.
Article in English | MEDLINE | ID: mdl-35550825

ABSTRACT

In 2007, it was shown that DNA sequence variants of the human NLRP1 gene are associated with autoimmune and autoinflammatory diseases affecting mainly the skin. However, at that time, the underlying cellular and molecular mechanisms were poorly characterized. Meanwhile, increasing evidence suggests that the NLRP1 inflammasome expressed by keratinocytes not only plays a part in the pathology of common inflammatory skin diseases and cancer development but also contributes to skin immunity. Understanding the mechanisms regulating NLRP1 activation in keratinocytes and the downstream events in human skin might pave the way for developing novel strategies for treating patients suffering from NLRP1-mediated skin diseases.


Subject(s)
Dermatitis , Skin Diseases , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Humans , Inflammasomes/metabolism , Keratinocytes/metabolism , NLR Proteins
3.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34884681

ABSTRACT

Apremilast (Otezla®) is an oral small molecule phosphodiesterase 4 (PDE4) inhibitor approved for the treatment of psoriasis, psoriatic arthritis, and oral ulcers associated with Behçet's disease. While PDE4 inhibition overall is mechanistically understood, the effect of apremilast on the innate immune response, particularly inflammasome activation, remains unknown. Here, we assessed the effect of apremilast in a psoriasis mouse model and primary human cells. Psoriatic lesion development in vivo was studied in K5.Stat3C transgenic mice treated with apremilast for 2 weeks, resulting in a moderate (2 mg/kg/day) to significant (6 mg/kg/day) resolution of inflamed plaques after 2-week treatment. Concomitantly, epidermal thickness dramatically decreased, the cutaneous immune cell infiltrate was reduced, and proinflammatory cytokines were significantly downregulated. Additionally, apremilast significantly inhibited lipopolysaccharide- or anti-CD3-induced expression of proinflammatory cytokines in peripheral mononuclear cells (PBMCs). Notably, inflammasome activation and secretion of IL-1ß were not inhibited by apremilast in PBMCs and in human primary keratinocytes. Collectively, apremilast effectively alleviated the psoriatic phenotype of K5.Stat3 transgenic mice, further substantiating PDE4 inhibitor-efficiency in targeting key clinical, histopathological and inflammatory features of psoriasis. Despite lacking direct effect on inflammasome activation, reduced priming of inflammasome components upon apremilast treatment reflected the indirect benefit of PDE4 inhibition in reducing inflammation.


Subject(s)
Interleukin-1beta/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , Psoriasis/drug therapy , Thalidomide/analogs & derivatives , Animals , Drug Evaluation, Preclinical , Humans , Immunity, Innate/drug effects , Inflammasomes/metabolism , Mice, Transgenic , Phosphodiesterase 4 Inhibitors/pharmacology , Psoriasis/metabolism , Thalidomide/pharmacology , Thalidomide/therapeutic use
4.
Biomedicines ; 9(7)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206503

ABSTRACT

p62 is a highly conserved, multi-domain, and multi-functional adaptor protein critically involved in several important cellular processes. Via its pronounced domain architecture, p62 binds to numerous interaction partners, thereby influencing key pathways that regulate tissue homeostasis, inflammation, and several common diseases including cancer. Via binding of ubiquitin chains, p62 acts in an anti-inflammatory manner as an adaptor for the auto-, xeno-, and mitophagy-dependent degradation of proteins, pathogens, and mitochondria. Furthermore, p62 is a negative regulator of inflammasome complexes. The transcription factor Nrf2 regulates expression of a bundle of ROS detoxifying genes. p62 activates Nrf2 by interaction with and autophagosomal degradation of the Nrf2 inhibitor Keap1. Moreover, p62 activates mTOR, the central kinase of the mTORC1 sensor complex that controls cell proliferation and differentiation. Through different mechanisms, p62 acts as a positive regulator of the transcription factor NF-κB, a central player in inflammation and cancer development. Therefore, p62 represents not only a cargo receptor for autophagy, but also a central signaling hub, linking several important pro- and anti-inflammatory pathways. This review aims to summarize knowledge about the molecular mechanisms underlying the roles of p62 in health and disease. In particular, different types of tumors are characterized by deregulated levels of p62. The elucidation of how p62 contributes to inflammation and cancer progression at the molecular level might promote the development of novel therapeutic strategies.

5.
Int J Mol Sci ; 21(13)2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32640751

ABSTRACT

Inflammasomes represent a group of protein complexes that contribute to host defense against pathogens and repair processes upon the induction of inflammation. However, aberrant and chronic inflammasome activation underlies the pathology of numerous common inflammatory diseases. Inflammasome assembly causes activation of the protease caspase-1 which in turn activates proinflammatory cytokines and induces a lytic type of cell death termed pyroptosis. Although NLRP1 (NACHT, leucine-rich repeat and pyrin domain containing 1) was the first inflammasome sensor, described almost 20 years ago, the molecular mechanisms underlying its activation and the resulting downstream events are incompletely understood. This is partially a consequence of the poor conservation of the NLRP1 pathway between human and mice. Moreover, recent evidence demonstrates a complex and multi-stage mechanism of NLRP1 inflammasome activation. In contrast to other inflammasome sensors, NLRP1 possesses protease activity required for proteolytic self-cleavage and activation mediated by the function-to-find domain (FIIND). CARD8 is a second FIIND protein and is expressed in humans but not in mice. In immune cells and AML (acute myeloid leukemia) cells, the anti-cancer drug talabostat induces CARD8 activation and causes caspase-1-dependent pyroptosis. In contrast, in human keratinocytes talabostat induces NLRP1 activation and massive proinflammatory cytokine activation. NLRP1 is regarded as the principal inflammasome sensor in human keratinocytes and UVB radiation induces its activation, which is believed to underlie the induction of sunburn. Moreover, gain-of-function mutations of NLRP1 cause inflammatory skin syndromes and a predisposition for the development of skin cancer. SNPs (single nucleotide polymorphisms) of NLRP1 are associated with several (auto)inflammatory diseases with a major skin phenotype, such as psoriasis or vitiligo. Here, we summarize knowledge about NLRP1 with emphasis on its role in human keratinocytes and skin. Due to its accessibility, pharmacological targeting of NLRP1 activation in epidermal keratinocytes represents a promising strategy for the treatment of the numerous patients suffering from NLRP1-dependent inflammatory skin conditions and cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Inflammasomes/metabolism , Inflammation/pathology , Keratinocytes/pathology , Skin Neoplasms/pathology , Skin/pathology , Animals , CARD Signaling Adaptor Proteins/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , NLR Proteins , Neoplasm Proteins/metabolism , Skin/immunology , Skin/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
6.
Biomolecules ; 10(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-32053878

ABSTRACT

The skin represents an indispensable barrier between the organism and the environment and is the first line of defense against exogenous insults. The transcription factor NRF2 is a central regulator of cytoprotection and stress resistance. NRF2 is activated in response to oxidative stress by reactive oxygen species (ROS) and electrophiles. These electrophiles oxidize specific cysteine residues of the NRF2 inhibitor KEAP1, leading to KEAP1 inactivation and, subsequently, NRF2 activation. As oxidative stress is associated with inflammation, the NRF2 pathway plays important roles in the pathogenesis of common inflammatory diseases and cancer in many tissues and organs, including the skin. The electrophile and NRF2 activator dimethyl fumarate (DMF) is an established and efficient drug for patients suffering from the common inflammatory skin disease psoriasis and the neuro-inflammatory disease multiple sclerosis (MS). In this review, we discuss possible molecular mechanisms underlying the therapeutic activity of DMF and other NRF2 activators. Recent evidence suggests that electrophiles not only activate NRF2, but also target other inflammation-associated pathways including the transcription factor NF-κB and the multi-protein complexes termed inflammasomes. Inflammasomes are central regulators of inflammation and are involved in many inflammatory conditions. Most importantly, the NRF2 and inflammasome pathways are connected at different levels, mainly antagonistically.


Subject(s)
Dermatologic Agents/pharmacology , Dimethyl Fumarate/pharmacology , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Skin Diseases/drug therapy , Skin Diseases/metabolism , Animals , Dermatologic Agents/therapeutic use , Dimethyl Fumarate/therapeutic use , Electrons , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism
7.
J Clin Invest ; 130(3): 1417-1430, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31805013

ABSTRACT

Epidermal growth factor receptor (EGFR) and MEK inhibitors (EGFRi/MEKi) are beneficial for the treatment of solid cancers but are frequently associated with severe therapy-limiting acneiform skin toxicities. The underlying molecular mechanisms are poorly understood. Using gene expression profiling we identified IL-36γ and IL-8 as candidate drivers of EGFRi/MEKi skin toxicity. We provide molecular and translational evidence that EGFRi/MEKi in concert with the skin commensal bacterium Cutibacterium acnes act synergistically to induce IL-36γ in keratinocytes and subsequently IL-8, leading to cutaneous neutrophilia. IL-36γ expression was the combined result of C. acnes-induced NF-κB activation and EGFRi/MEKi-mediated expression of the transcription factor Krüppel-like factor 4 (KLF4), due to the presence of both NF-κB and KLF4 binding sites in the human IL-36γ gene promoter. EGFRi/MEKi increased KLF4 expression by blockade of the EGFR/MEK/ERK pathway. These results provide an insight into understanding the pathological mechanism of the acneiform skin toxicities induced by EGFRi/MEKi and identify IL-36γ and the transcription factor KLF4 as potential therapeutic targets.


Subject(s)
ErbB Receptors/immunology , Interleukin-1/immunology , MAP Kinase Signaling System/immunology , Propionibacteriaceae/immunology , Skin Diseases, Bacterial/immunology , Animals , ErbB Receptors/genetics , Humans , Interleukin-1/genetics , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Skin Diseases, Bacterial/genetics , Skin Diseases, Bacterial/pathology
8.
J Invest Dermatol ; 140(7): 1335-1345.e10, 2020 07.
Article in English | MEDLINE | ID: mdl-31877317

ABSTRACT

Inflammasome activation induces caspase-1-dependent secretion of the proinflammatory cytokine IL-1ß. In addition, caspase-1 activates the protein GSDMD in immune cells, causing pyroptosis, a lytic type of cell death. In contrast, UVB irradiation of human primary keratinocytes induces NLRP1 inflammasome activation, cytokine secretion, and caspase-1-dependent apoptosis, rather than pyroptosis. Here, we addressed the molecular mechanisms underlying the role of caspase-1 in UVB-induced cell death of human primary keratinocytes. We show that GSDMD is a poor substrate of caspase-1 in human primary keratinocytes and that its activation upon UVB irradiation supports secretion of IL-1ß. We screened for novel substrates of caspase-1 by a mass spectrometry-based approach and identified the specific cleavage of the major vault protein (MVP) at D441 by caspase-1 and -9. MVP is the main component of vaults, highly conserved ribonucleoprotein particles, whose functions are poorly understood. Cleavage of MVP is a common event occurring in human primary keratinocytes and fibroblasts undergoing apoptosis induced by different stimuli. In contrast, MVP cleavage could not be detected in pyroptotic cells. Cleavage of MVP by caspase-1 and -9 inactivates this cytoprotective protein. These results demonstrate a proapoptotic activity of caspase-1 and a crosstalk with caspase-9 upon inactivation of the cytoprotective MVP in apoptotic epithelial cells.


Subject(s)
Apoptosis , Caspase 1/metabolism , Caspase 9/metabolism , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Vault Ribonucleoprotein Particles/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Biopsy , Fibroblasts/metabolism , Humans , Inflammasomes , Interleukin-1beta/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mass Spectrometry , NLR Proteins , RNA, Small Interfering/metabolism , Ultraviolet Rays
9.
Methods Mol Biol ; 2109: 125-145, 2020.
Article in English | MEDLINE | ID: mdl-31502220

ABSTRACT

The culture of epidermal human primary keratinocytes (HPKs) represents a well-established model in biological and dermatological research. In addition, HPKs are used in three-dimensional organotypic cultures (OTCs), and gene therapeutic approaches have been reported for the treatment of patients suffering from epidermolysis bullosa, a severe blistering disease that can result in postnatal lethality. Therefore, there is a strong need for the development of techniques for the stable and specific genetic manipulation of HPKs, for example, by genome editing via the CRISPR/Cas9 approach. However, the main disadvantage of working with HPKs is the fact that these cells are prone to terminal differentiation and proliferate only for few passages in monoculture. As it is well known that the co-culture of HPKs with fibroblasts strongly increases the lifetime of the epidermal cells, we developed a protocol for the stable modification of HPKs by CRISPR/Cas9 via lentiviral transduction in the presence of 3T3-J2 fibroblasts as feeder cells. Selection of transduced HPKs is achieved with antibiotics in co-culture with antibiotic-resistant feeder cells. Modified HPKs generated by our protocol have the potential to generate epidermis-like structures in OTCs.


Subject(s)
Feeder Cells/cytology , Gene Editing/methods , Gene Knockout Techniques/methods , Keratinocytes/cytology , 3T3-L1 Cells , Animals , CRISPR-Cas Systems , Cells, Cultured , Gene Expression , Humans , Keratinocytes/metabolism , Mice , Primary Cell Culture
10.
J Invest Dermatol ; 139(8): 1788-1797.e6, 2019 08.
Article in English | MEDLINE | ID: mdl-30738816

ABSTRACT

The inflammasome protein NLRP1 is an important innate immune sensor in human keratinocytes, and, together with ASC and caspase-1, it mediates the activation and secretion of the proinflammatory cytokines IL-1ß and IL-18. These cytokines and inflammasomes can have partly opposing roles during tumorigenesis in mice. In contrast, ASC expression is impaired in different types of cancer in humans. In this study, we analyzed inflammasome activation and expression of inflammasome proteins, including their downstream cytokines, in squamous cell carcinomas, a type of nonmelanoma skin cancer derived from keratinocytes. We assessed mRNA and protein levels in human primary keratinocytes and skin carcinoma-derived SCC cell lines and detected a strong down-regulation of expression of NLRP1 inflammasome components, as well as reduced expression of the proinflammatory cytokines proIL-1ß and proIL-1α. Protein levels of NLRP1, ASC, caspase-1, and proIL-1ß were reduced in patient-derived SCC biopsy samples compared with healthy skin. Furthermore, the results suggest that expression of PYCARD (ASC), CASP1, IL1B, and NLRP1 is silenced by methylation in SCC cell lines. In conclusion, the down-regulation of the inflammasome pathway in SCCs might favor late tumor development in human skin.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic/immunology , Inflammasomes/genetics , Skin Neoplasms/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Biopsy , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cytokines , Down-Regulation , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Keratinocytes/immunology , Keratinocytes/pathology , NLR Proteins , Primary Cell Culture , RNA, Messenger/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Skin/cytology , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology
11.
J Invest Dermatol ; 138(12): 2644-2652, 2018 12.
Article in English | MEDLINE | ID: mdl-30096351

ABSTRACT

By forming a protective barrier, epidermal keratinocytes represent the first line of defense against environmental insults. UVB radiation of the sun is a major challenge for the skin and can induce inflammation, aging, and eventually skin cancer. UVB induces an immune response in human keratinocytes resulting in activation and secretion of the proinflammatory cytokines proIL-1ß and -18. This is mediated by an assembly of protein complexes, termed inflammasomes. However, the mechanisms underlying sensing of UVB by keratinocytes, and particularly the types of inflammasomes required for cytokine secretion, are a matter of debate. To address these questions, we established a protocol that allows the generation of CRISPR/Cas9-targeted human primary keratinocytes. Our experiments showed an essential role of the NLRP1 rather than the NLRP3 inflammasome in UVB sensing and subsequent IL-1ß and -18 secretion by keratinocytes. Moreover, NLRP1 but not NLRP3 was required for inflammasome activation in response to nigericin, a potassium ionophore and well-established NLRP3 activator in immune cells. Because the CRISPR/Cas9-targeted cells retained their full differentiation capacity, genome editing of human primary keratinocytes might be useful for numerous research and medical applications.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CRISPR-Cas Systems , Gene Editing/methods , Inflammasomes/metabolism , Keratinocytes/physiology , Radiodermatitis/genetics , Ultraviolet Rays/adverse effects , CRISPR-Associated Protein 9/genetics , Cells, Cultured , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins , Nigericin/pharmacology , Primary Cell Culture , Radiodermatitis/metabolism
12.
J Immunol ; 200(9): 3151-3159, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29592962

ABSTRACT

Allergen immunotherapy (AIT) is the only modality that can modify immune responses to allergen exposure, but therapeutic coverage is low. One strategy to improve AIT safety and efficacy is the use of new or improved adjuvants. This study investigates immune responses produced by microcrystalline tyrosine (MCT)-based vaccines as compared with conventional aluminum hydroxide (alum). Wild-type, immune-signaling-deficient, and TCR-transgenic mice were treated with different Ags (e.g., OVA and cat dander Fel d 1), plus MCT or alum as depot adjuvants. Specific Ab responses in serum were measured by ELISA, whereas cytokine secretion was measured both in culture supernatants by ELISA or by flow cytometry of spleen cells. Upon initiation of AIT in allergic mice, body temperature and further clinical signs were used as indicators for anaphylaxis. Overall, MCT and alum induced comparable B and T cell responses, which were independent of TLR signaling. Alum induced stronger IgE and IL-4 secretion than MCT. MCT and alum induced caspase-dependent IL-1ß secretion in human monocytes in vitro, but inflammasome activation had no functional effect on inflammatory and Ab responses measured in vivo. In sensitized mice, AIT with MCT-adjuvanted allergens caused fewer anaphylactic reactions compared with alum-adjuvanted allergens. As depot adjuvants, MCT and alum are comparably effective in strength and mechanism of Ag-specific IgG induction and induction of T cell responses. The biocompatible and biodegradable MCT seems therefore a suitable alternative adjuvant to alum-based vaccines and AIT.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Desensitization, Immunologic/methods , Tyrosine/pharmacology , Animals , Disease Models, Animal , Hypersensitivity/prevention & control , Immunoglobulin E/immunology , Inflammasomes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology , Toll-Like Receptors/immunology
13.
J Invest Dermatol ; 138(6): 1380-1390, 2018 06.
Article in English | MEDLINE | ID: mdl-29287762

ABSTRACT

Inflammasomes are key intracellular signaling platforms involved in innate immune responses to micro-organisms and danger signals. Extracellular signal-regulated kinase, Jun N-terminal kinase, and p38 mitogen-activated protein kinase family members are activated by numerous environmental stresses. Recently, it has been reported that Jun N-terminal kinase is involved in inflammasome activation in myeloid immune cells. To date, the role of mitogen-activated protein kinase in inflammasome activity in keratinocytes has not been investigated. Here, we show that, in primary human keratinocytes, p38 mitogen-activated protein kinase is required for inflammasome activation and IL-1ß secretion. Using selective small molecule inhibitors, small interfering RNA gene silencing, and CRISPR/Cas9-based deletion, we demonstrate the above and identify p38α and p38δ as critical regulators of ASC oligomerization, inflammasome activation, and IL-1ß secretion in keratinocytes. Furthermore, our data suggest that the nature of the mitogen-activated protein kinase regulating inflammasome activity exhibits a certain cell specificity, with p38 playing a predominant role in keratinocytes and Jun N-terminal kinase 1 in cells of myeloid origin.


Subject(s)
Inflammasomes/immunology , Keratinocytes/immunology , Mitogen-Activated Protein Kinase 13/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Signal Transduction/immunology , CARD Signaling Adaptor Proteins/immunology , CARD Signaling Adaptor Proteins/metabolism , CRISPR-Cas Systems/genetics , Cells, Cultured , Enzyme Activation , Humans , Inflammasomes/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases , Keratinocytes/metabolism , Mitogen-Activated Protein Kinase 13/antagonists & inhibitors , Mitogen-Activated Protein Kinase 13/genetics , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phosphorylation , Primary Cell Culture , Protein Multimerization/immunology , RNA Interference , RNA, Small Interfering/metabolism
14.
Front Pharmacol ; 8: 278, 2017.
Article in English | MEDLINE | ID: mdl-28588486

ABSTRACT

In 2002, intracellular protein complexes known as the inflammasomes were discovered and were shown to have a crucial role in the sensing of intracellular pathogen- and danger-associated molecular patterns (PAMPs and DAMPs). Activation of the inflammasomes results in the processing and subsequent secretion of the pro-inflammatory cytokines IL-1ß and IL-18. Several autoinflammatory disorders such as cryopyrin-associated periodic syndromes and Familial Mediterranean Fever have been associated with mutations of genes encoding inflammasome components. Moreover, the importance of IL-1 has been reported for an increasing number of autoinflammatory skin diseases including but not limited to deficiency of IL-1 receptor antagonist, mevalonate kinase deficiency and PAPA syndrome. Recent findings have revealed that excessive IL-1 release induced by harmful stimuli likely contributes to the pathogenesis of common dermatological diseases such as acne vulgaris or seborrheic dermatitis. A key pathogenic feature of these diseases is IL-1ß-induced neutrophil recruitment to the skin. IL-1ß blockade may therefore represent a promising therapeutic approach. Several case reports and clinical trials have demonstrated the efficacy of IL-1 inhibition in the treatment of these skin disorders. Next to the recombinant IL-1 receptor antagonist (IL-1Ra) Anakinra and the soluble decoy Rilonacept, the anti-IL-1α monoclonal antibody MABp1 and anti-IL-1ß Canakinumab but also Gevokizumab, LY2189102 and P2D7KK, offer valid alternatives to target IL-1. Although less thoroughly investigated, an involvement of IL-18 in the development of cutaneous inflammatory disorders is also suspected. The present review describes the role of IL-1 in diseases with skin involvement and gives an overview of the relevant studies discussing the therapeutic potential of modulating the secretion and activity of IL-1 and IL-18 in such diseases.

15.
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
16.
Sci Rep ; 6: 29914, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27426915

ABSTRACT

Hypoxia is a hallmark of cancer that is strongly associated with invasion, metastasis, resistance to therapy and poor clinical outcome. Tumour hypoxia affects immune responses and promotes the accumulation of macrophages in the tumour microenvironment. However, the signals linking tumour hypoxia to tumour-associated macrophage recruitment and tumour promotion are incompletely understood. Here we show that the damage-associated molecular pattern High-Mobility Group Box 1 protein (HMGB1) is released by melanoma tumour cells as a consequence of hypoxia and promotes M2-like tumour-associated macrophage accumulation and an IL-10 rich milieu within the tumour. Furthermore, we demonstrate that HMGB1 drives IL-10 production in M2-like macrophages by selectively signalling through the Receptor for Advanced Glycation End products (RAGE). Finally, we show that HMGB1 has an important role in murine B16 melanoma growth and metastasis, whereas in humans its serum concentration is significantly increased in metastatic melanoma. Collectively, our findings identify a mechanism by which hypoxia affects tumour growth and metastasis in melanoma and depict HMGB1 as a potential therapeutic target.


Subject(s)
Macrophages/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Tumor Hypoxia , Animals , Cell Proliferation , Female , HMGB1 Protein/blood , HMGB1 Protein/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , Macrophages/pathology , Male , Melanoma, Experimental/blood , Mice, Inbred C57BL , Phenotype , Receptor for Advanced Glycation End Products/metabolism , Skin Neoplasms/blood , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
17.
Exp Dermatol ; 23(12): 884-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267545

ABSTRACT

Although being a normal part of the skin flora, yeasts of the genus Malassezia are associated with several common dermatologic conditions including pityriasis versicolour, seborrhoeic dermatitis (SD), folliculitis, atopic eczema/dermatitis (AE/AD) and dandruff. While Malassezia spp. are aetiological agents of pityriasis versicolour, a causal role of Malassezia spp. in AE/AD and SD remains to be established. Previous reports have shown that fungi such as Candida albicans and Aspergillus fumigatus are able to efficiently activate the NLRP3 inflammasome leading to robust secretion of the pro-inflammatory cytokine IL-1ß. To date, innate immune responses to Malassezia spp. are not well characterized. Here, we show that different Malassezia species could induce NLRP3 inflammasome activation and subsequent IL-1ß secretion in human antigen-presenting cells. In contrast, keratinocytes were not able to secrete IL-1ß when exposed to Malassezia spp. Moreover, we demonstrate that IL-1ß secretion in antigen-presenting cells was dependent on Syk-kinase signalling. Our results identify Malassezia spp. as potential strong inducers of pro-inflammatory responses when taken up by antigen-presenting cells and identify C-type lectin receptors and the NLRP3 inflammasome as crucial actors in this process.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/microbiology , Carrier Proteins/immunology , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Malassezia/immunology , Protein-Tyrosine Kinases/metabolism , Animals , Antigen-Presenting Cells/metabolism , Carrier Proteins/genetics , Caspase 1/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Dermatomycoses/immunology , Dermatomycoses/metabolism , Dermatomycoses/microbiology , Humans , Immunity, Innate , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Lectins, C-Type/metabolism , Malassezia/genetics , Malassezia/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Syk Kinase
19.
J Invest Dermatol ; 134(3): 677-685, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24157462

ABSTRACT

Acne vulgaris is potentially a severe skin disease associated with colonization of the pilo-sebaceous unit by the commensal bacterium Propionibacterium acnes and inflammation. P. acnes is considered to contribute to inflammation in acne, but the pathways involved are unclear. Here we reveal a mechanism that regulates inflammatory responses to P. acnes. We show that IL-1ß mRNA and the active processed form of IL-1ß are abundant in inflammatory acne lesions. Moreover, we identify P. acnes as a trigger of monocyte-macrophage NLRP3-inflammasome activation, IL-1ß processing and secretion that is dependent on phagocytosis, lysosomal destabilization, reactive oxygen species, and cellular K+ efflux. In mice, inflammation induced by P. acnes is critically dependent on IL-1ß and the NLRP3 inflammasome of myeloid cells. These findings show that the commensal P. acnes-by activating the inflammasome-can trigger an innate immune response in the skin, thus establishing the NLRP3-inflammasome and IL-1ß as possible therapeutic targets in acne.


Subject(s)
Acne Vulgaris/immunology , Gram-Positive Bacterial Infections/immunology , Interleukin-1beta/immunology , Monocytes/immunology , Monocytes/microbiology , Propionibacterium acnes/immunology , Acne Vulgaris/metabolism , Acne Vulgaris/microbiology , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Gram-Positive Bacterial Infections/metabolism , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Keratinocytes/cytology , Keratinocytes/immunology , Keratinocytes/microbiology , Leukemia , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...