1
|
Huang YT, Chiu LY, Lu PH, Hsiao PF, Wang JY, Lu PH, Wu NL. AMPK activation modulates IL-36-induced inflammatory responses by regulating IκBζ expression in the skin. Br J Pharmacol 2024; 181:2429-2442. [PMID: 38532634 DOI: 10.1111/bph.16354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 12/27/2023] [Accepted: 01/21/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND AND PURPOSE The interleukin (IL)-36 pathway is a critical player in the pathogenesis of pustular psoriasis. However, therapies targeting this pathway are limited or unaffordable (e.g. the anti-IL-36 receptor antibody). AMP-activated protein kinase (AMPK), a regulator of cellular energy and metabolism, is known to participate in inflammatory diseases. However, its role in IL-36-induced skin inflammation remains unclear. Therefore, we sought to investigate the role of AMPK signals in regulating IL-36-induced responses in the skin. EXPERIMENTAL APPROACH IL-36-stimulated primary normal human epidermal keratinocytes (NHEKs) and IL-36-injected (intradermally) BALB/c mice served as the cell and animal models, respectively. Additionally, 5-aminoimidazole-4-carboxamide riboside (AICAR) and A769662 served as AMPK activators. KEY RESULTS AICAR and A769662 significantly suppressed the IL-36-induced IL-8 (CXCL8) and CCL20 production from NHEKs. IL-36-induced IκBζ protein expression was prominently reduced and IKK/IκBα phosphorylation was attenuated by AICAR and A769662. Conversely, AMPKα knockdown increased IκBζ protein expression and IKK/IκBα phosphorylation in IL-36-treated NHEKs. Furthermore, AICAR and A769662 enhanced IL-36-induced-IκBζ protein degradation via the proteasome-dependent but not the lysosome-dependent pathway. Pretreatment of NHEKs with IL-36 slightly suppressed the AICAR- and A769662-triggered phosphorylation of AMPK and acetyl-CoA carboxylase. In the mouse model, topical application of AICAR significantly reduced ear swelling, redness, epidermal thickening, neutrophil infiltration and inflammatory and antimicrobial peptide gene expression. CONCLUSION AND IMPLICATIONS AMPK activation suppresses IL-36-induced IL-8 and CCL20 release by regulating IκBζ expression in keratinocytes and reduces IL-36-induced skin inflammation in mice, suggesting that AMPK activation is a potential strategy for treating patients with IL-36-mediated inflammatory skin disorders.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ling-Ya Chiu
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Nursing, MacKay Medical College, New Taipei City, Taiwan
| | - Po-Hsuan Lu
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Pa-Fan Hsiao
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Jen-Yu Wang
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Ping-Hsun Lu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Nan-Lin Wu
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
| |
Collapse
|
2
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 DOI: 10.3390/cells13060521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d'Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
3
|
Fernández-Lázaro D, Sanz B, Seco-Calvo J. The Mechanisms of Regulated Cell Death: Structural and Functional Proteomic Pathways Induced or Inhibited by a Specific Protein-A Narrative Review. Proteomes 2024; 12:3. [PMID: 38250814 PMCID: PMC10801515 DOI: 10.3390/proteomes12010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Billions of cells die in us every hour, and our tissues do not shrink because there is a natural regulation where Cell Death (CD) is balanced with cell division. The process in which cells eliminate themselves in a controlled manner is called Programmed Cell Death (PCD). The PCD plays an important role during embryonic development, in maintaining homeostasis of the body's tissues, and in the elimination of damaged cells, under a wide range of physiological and developmental stimuli. A multitude of protein mediators of PCD have been identified and signals have been found to utilize common pathways elucidating the proteins involved. This narrative review focuses on caspase-dependent and caspase-independent PCD pathways. Included are studies of caspase-dependent PCD such as Anoikis, Catastrophe Mitotic, Pyroptosis, Emperitosis, Parthanatos and Cornification, and Caspase-Independent PCD as Wallerian Degeneration, Ferroptosis, Paraptosis, Entosis, Methuosis, and Extracellular Trap Abnormal Condition (ETosis), as well as neutrophil extracellular trap abnormal condition (NETosis) and Eosinophil Extracellular Trap Abnormal Condition (EETosis). Understanding PCD from those reported in this review could shed substantial light on the processes of biological homeostasis. In addition, identifying specific proteins involved in these processes is mandatory to identify molecular biomarkers, as well as therapeutic targets. This knowledge could provide the ability to modulate the PCD response and could lead to new therapeutic interventions in a wide range of diseases.
Collapse
Affiliation(s)
- Diego Fernández-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
| | - Begoña Sanz
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Jesús Seco-Calvo
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Institute of Biomedicine (IBIOMED), Universidad de León, 27071 León, Spain
| |
Collapse
|
4
|
Hsiao PF, Huang YT, Lu PH, Chiu LY, Weng TH, Hung CF, Wu NL. Thioredoxin-interacting protein regulates keratinocyte differentiation: Implication of its role in psoriasis. FASEB J 2022; 36:e22313. [PMID: 35471587 DOI: 10.1096/fj.202101772r] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022]
Abstract
Thioredoxin-interacting protein (TXNIP), also known as Vitamin-D upregulated protein-1 (VDUP-1), interacts with thioredoxin to regulate redox responses and participates in diverse disorders including metabolic, cardiovascular, inflammatory and malignant diseases. Psoriasis is characterized by chronic skin inflammation and an aberrant pattern of keratinocyte differentiation. Clinically, psoriasis is associated with various cardiometabolic comorbidities but studies on TXNIP's biological role in skin disorders are limited. In this study, we investigated TXNIP expression in psoriasis and its regulation in normal human epidermal keratinocytes (NHEKs), and then explored how TXNIP regulated skin keratinocyte differentiation to determine its role in psoriasis pathogenesis. Our immunohistochemical study demonstrated extensive TXNIP expression in the upper and lower epidermis of psoriasis compared to predominant TXNIP expression in the basal layer of normal skin. 1, 25-dihydroxyvitamin D3 suppressed but TGF-α and EGF enhanced TXNIP expression in NHEKs. An inducer of keratinocyte differentiation, phorbol 12-myristate 13-acetate (PMA), also diminished TXNIP expression, which was reversed by PKC-δ knockdown. TXNIP knockdown reduced PMA-induced involucrin and transglutaminse-1 expression, and increased p63 expression in NHEKs but did not significantly affect cell proliferation. H2 O2 -induced ROS production and EGFR phosphorylation decreased in NHEKs with TXNIP knockdown. Furthermore, PMA-induced PKC-δ phosphorylation, TGF-α, and EGF-triggered EGFR phosphorylation were attenuated by TXNIP knockdown. Our results unraveled the regulation and function of TXNIP expression in skin keratinocytes and the cross-regulation between TXNIP and EGFR signaling. These findings imply a role of TXNIP in psoriasis and provide insight into the possible impact of TXNIP regulators on the skin or psoriasis.
Collapse
Affiliation(s)
- Pa-Fan Hsiao
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Yi-Ting Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Po-Hsuan Lu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Ling-Ya Chiu
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Han Weng
- Department of Medical Education, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.,Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
| |
Collapse
|
5
|
Wu NL, Huang DY, Hsieh SL, Dai YS, Lin WW. Decoy receptor 3 is involved in epidermal keratinocyte commitment to terminal differentiation via EGFR and PKC activation. Exp Mol Med 2022; 54:542-551. [PMID: 35478210 PMCID: PMC9076855 DOI: 10.1038/s12276-022-00762-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/04/2021] [Accepted: 01/09/2022] [Indexed: 11/30/2022] Open
Abstract
Decoy receptor 3 (DcR3) is a soluble receptor for Fas ligand, LIGHT and TL1A, but it also exerts effector functions. Previously, we found that DcR3 is upregulated in the serum and lesional skin of patients with psoriasis and is upregulated by EGFR activation in proliferating primary human epidermal keratinocytes. However, the functional role of intracellular DcR3 in keratinocyte differentiation is still incompletely defined. Herein, primary cultured human epidermal keratinocytes were differentiated by phorbol 12-myristate 13-acetate (PMA) treatment, calcium treatment and cell confluence, which are three standard in vitro differentiation models. We found that the constitutive expression of the DcR3 gene and protein was progressively suppressed during terminal differentiation of keratinocytes. These changes were correlated with downregulation of EGFR activation during keratinocyte differentiation. EGFR inhibition by gefitinib further decreased confluence-induced suppression of DcR3 mRNA expression, and, vice versa, knocking down DcR3 expression attenuated EGFR and EGFR ligand expression as well as EGFR activation. Under conditions without a change in cell growth, DcR3 silencing reduced the expression of involucrin and transglutaminase 1 but enhanced the induction of the terminal differentiation markers keratin 10 and loricrin. Of note, DcR3 interacted with PKCα and PKCδ and enhanced PKC activity. In keratinocytes with PKCα and PKCδ silencing, differentiation markers were differentially affected. In conclusion, DcR3 expression in keratinocytes is regulated by EGFR and forms a positive feedback loop to orchestrate constitutive EGFR and PKC activity. During differentiation, DcR3 is downregulated and involved in modulating the pattern of terminal differentiation. A protein linked to cancer and various inflammatory diseases may also be an important driver for the skin condition in psoriasis. The outer surface of the skin is formed by cells called keratinocytes, which transition from a highly proliferative state to a fully mature state where they no longer divide. This developmental process is disrupted in psoriasis. Researchers led by Wan-Wan Lin at National Taiwan University, Taipei, have now identified a prominent role for a protein called decoy receptor 3 (DcR3), which is a biomarker for a variety of disorders and is also abnormally expressed in keratinocytes in psoriatic lesions. Lin and colleagues demonstrated that DcR3 interacts with multiple cellular signaling pathways that coordinate cell differentiation. These findings reveal how aberrant DcR3 activity might lead to the abnormal keratinocyte developmental behavior observed in psoriasis.
Collapse
Affiliation(s)
- Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan, ROC.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan, ROC.,Institute of Biomedical Sciences, Mackay Medical College, New Taipei, Taiwan, ROC
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | | | - Yang-Shia Dai
- Department of Dermatology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC. .,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan, ROC. .,Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
6
|
Man MQ, Wakefield JS, Mauro TM, Elias PM. Role of nitric oxide in regulating epidermal permeability barrier function. Exp Dermatol 2022; 31:290-298. [PMID: 34665906 PMCID: PMC8897205 DOI: 10.1111/exd.14470] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO), a free radical molecule synthesized by nitric oxide synthases (NOS), regulates multiple cellular functions in a variety of cell types. These NOS, including endothelial NOS (eNOS), inducible NOS (iNOS) and neural NOS (nNOS), are expressed in keratinocytes. Expression levels of both iNOS and nNOS decrease with ageing, and insufficient NO has been linked to the development of a number of disorders such as diabetes and hypertension, and to the severity of atherosclerosis. Conversely, excessive NO levels can induce cellular oxidative stress, but physiological levels of NO are required to maintain the normal functioning of cells, including keratinocytes. NO also regulates cutaneous functions, including epidermal permeability barrier homeostasis and wound healing, through its stimulation of keratinocyte proliferation, differentiation and lipid metabolism. Topical applications of a diverse group of agents which generate nitric oxide (called NO donors) such as S-nitroso-N-acetyl-D,L-penicillamine (SNAP) can delay permeability barrier recovery in barrier-disrupted skin, but iNOS is still required for epidermal permeability barrier homeostasis. This review summarizes the regulatory role that NO plays in epidermal permeability barrier functions and the underlying mechanisms involved.
Collapse
Affiliation(s)
- Mao-Qiang Man
- Dermatology Service, Veterans Affairs Medical Center San Francisco, and Department of Dermatology, University of California San Francisco, CA, USA,Dermatology Hospital, Southern Medical University, Guangdong 510091, China
| | - Joan S. Wakefield
- Dermatology Service, Veterans Affairs Medical Center San Francisco, and Department of Dermatology, University of California San Francisco, CA, USA
| | - Theodora M. Mauro
- Dermatology Service, Veterans Affairs Medical Center San Francisco, and Department of Dermatology, University of California San Francisco, CA, USA
| | - Peter M. Elias
- Dermatology Service, Veterans Affairs Medical Center San Francisco, and Department of Dermatology, University of California San Francisco, CA, USA
| |
Collapse
|
7
|
Lu PH, Wang JY, Chiu LY, Huang YT, Hung CF, Wu NL. Spleen tyrosine kinase regulates keratinocyte inflammasome activation and skin inflammation induced by UVB irradiation. Free Radic Biol Med 2022; 180:121-133. [PMID: 35007704 DOI: 10.1016/j.freeradbiomed.2022.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/09/2021] [Accepted: 01/05/2022] [Indexed: 12/31/2022]
Abstract
UVB can induce inflammatory responses contributing to diverse skin damage. UVB-triggered inflammasome activation of human keratinocytes underlies UVB-induced skin sunburn reaction. Pleiotropic functions of spleen tyrosine kinase (Syk) have rendered it as a potential therapeutic target. In immunocytes, Syk modulates immunoreceptor signaling and NLRP3 inflammasome activation. In skin, Syk mediates EGFR signaling, regulates keratinocyte differentiation and is involved in inflammatory disorders. However, roles of Syk in UVB-induced inflammasome activation in keratinocytes remain elusive. We investigated roles of keratinocyte Syk in UVB-triggered photo-responses. Primary normal human epidermal keratinocytes (NHEKs) isolated from skin were used. Syk knockdown or Syk inhibitor R406 was applied to investigate functions of keratinocyte Syk in UVB photobiology. The possible in vivo role of Syk was evaluated by checking UVB-induced skin damage in R406-treated mice. UVB was able to induce Syk phosphorylation in NHEKs that could be regulated by reactive oxygen species (ROS) generation and EGFR. Syk knockdown or Syk inhibitor (R406) treatment reduced UVB-triggered apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) crosslinking, procaspase-1 cleavage, active IL-1β formation, and gasdermin D activation, indicating roles of Syk in UVB-triggered inflammasome activation in keratinocytes. UVB-induced production of IL-8, TNF-α, ROS, and phosphorylation of JNK and p38 were attenuated after Syk knockdown or inhibition. R406 ameliorated UVB-induced mouse skin damage, including erythema and transepidermal water loss (TEWL). Thus, Syk participated in UVB-induced inflammasome activation and inflammatory response in vitro and in vivo, suggesting potential photo-protective effects of Syk inhibition in UVB-induced skin inflammation.
Collapse
Affiliation(s)
- Po-Hsuan Lu
- Department of Dermatology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei City, 10449, Taiwan; Department of Medicine, MacKay Medical College, No.46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 25245, Taiwan.
| | - Jen-Yu Wang
- Department of Dermatology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei City, 10449, Taiwan; MacKay Junior College of Medicine, Nursing, and Management, No. 42, Sec. 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 25245, Taiwan.
| | - Ling-Ya Chiu
- Department of Medical Research, MacKay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan; Department of Pharmacology, College of Medicine, National Taiwan University, No.1 Jen Ai Road Section 1, Taipei, 100233, Taiwan.
| | - Yi-Ting Huang
- Department of Medicine, MacKay Medical College, No.46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 25245, Taiwan; Department of Medical Research, MacKay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan.
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 242062, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei City, 10449, Taiwan; Department of Medicine, MacKay Medical College, No.46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 25245, Taiwan; MacKay Junior College of Medicine, Nursing, and Management, No. 42, Sec. 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 25245, Taiwan; Institute of Biomedical Sciences, MacKay Medical College, New Taipei, Taiwan.
| |
Collapse
|
8
|
Braegelmann C, Niebel D, Ferring-Schmitt S, Fetter T, Landsberg J, Hölzel M, Effern M, Glodde N, Steinbuch S, Bieber T, Wenzel J. Epigallocatechin-3-gallate exhibits anti-inflammatory effects in a human interface dermatitis model-implications for therapy. J Eur Acad Dermatol Venereol 2021; 36:144-153. [PMID: 34585800 DOI: 10.1111/jdv.17710] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/15/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epigallocatechin-3-gallate (EGCG) has been proven effective in treating viral warts. Since anticarcinogenic as well as anti-inflammatory properties are ascribed to the substance, its use has been evaluated in the context of different dermatoses. The effect of EGCG on interface dermatitis (ID), however, has not yet been explored. OBJECTIVES In this study, we investigated the effect of EGCG on an epidermal human in vitro model of ID. METHODS Via immunohistochemistry, lesional skin of lichen planus patients and healthy skin were analysed concerning the intensity of interferon-associated mediators, CXCL10 and MxA. Epidermal equivalents were stained analogously upon ID-like stimulation and EGCG treatment. Monolayer keratinocytes were treated likewise and supernatants were analysed via ELISA while cells were processed for vitality assay or transcriptomic analysis. RESULTS CXCL10 and MxA are strongly expressed in lichen planus lesions and induced in keratinocytes upon ID-like stimulation. EGCG reduces CXCL10 and MxA staining intensity in epidermis equivalents and CXCL10 secretion by keratinocytes upon stimulation. It furthermore minimizes the cytotoxic effect of the stimulus and downregulates a magnitude of typical pro-inflammatory cytokines that are crucial for the perpetuation of ID. CONCLUSIONS We provide evidence concerning anti-inflammatory effects of EGCG within a human in vitro model of ID. The capacity to suppress mediators that are centrally involved in disease perpetuation suggests EGCG as a potential topical therapeutic in lichen planus and other autoimmune skin diseases associated with ID.
Collapse
Affiliation(s)
- C Braegelmann
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - D Niebel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - S Ferring-Schmitt
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - T Fetter
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - J Landsberg
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - M Hölzel
- Institute of Experimental Oncology (IEO), University Hospital Bonn, Bonn, Germany
| | - M Effern
- Institute of Experimental Oncology (IEO), University Hospital Bonn, Bonn, Germany
| | - N Glodde
- Institute of Experimental Oncology (IEO), University Hospital Bonn, Bonn, Germany
| | - S Steinbuch
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - T Bieber
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - J Wenzel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
9
|
Qiu XM, Jiang L. [New diagnostic classification of cheilitis and its clinical diagnostic pathway]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:238-244. [PMID: 33834683 DOI: 10.7518/hxkq.2021.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cheilitis is a general term for various types of inflammatory diseases that occur on the lips. The etiology differs and the clinical manifestations and pathological features overlap, leading to difficulties in clinical diagnosis. Reasonable classification is conducive to the diagnosis of cheilitis. However, its classification is difficult because of its cha-racteristics. At present, scholars have proposed two different classification schemes, but a unified classification standard has not yet been established. We classified cheilitis based on its etiology, clinical manifestations, pathological characteristics, and relationship with systemic and special diseases on the basis of special medical reports and by combining clinical practice experience and summarizing previous cheilitis diagnosis and treatment literature. In accordance with this classification method, we proposed suggestions for the clinical diagnosis of cheilitis to provide a reference for the clinical diagnosis and treatment of complex cheilitis.
Collapse
Affiliation(s)
- Xue-Mei Qiu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Peyre L, Meyer M, Hofman P, Roux J. TRAIL receptor-induced features of epithelial-to-mesenchymal transition increase tumour phenotypic heterogeneity: potential cell survival mechanisms. Br J Cancer 2021; 124:91-101. [PMID: 33257838 PMCID: PMC7782794 DOI: 10.1038/s41416-020-01177-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
The continuing efforts to exploit the death receptor agonists, such as the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), for cancer therapy, have largely been impaired by the anti-apoptotic and pro-survival signalling pathways leading to drug resistance. Cell migration, invasion, differentiation, immune evasion and anoikis resistance are plastic processes sharing features of the epithelial-to-mesenchymal transition (EMT) that have been shown to give cancer cells the ability to escape cell death upon cytotoxic treatments. EMT has recently been suggested to drive a heterogeneous cellular environment that appears favourable for tumour progression. Recent studies have highlighted a link between EMT and cell sensitivity to TRAIL, whereas others have highlighted their effects on the induction of EMT. This review aims to explore the molecular mechanisms by which death signals can elicit an increase in response heterogeneity in the metastasis context, and to evaluate the impact of these processes on cell responses to cancer therapeutics.
Collapse
Affiliation(s)
- Ludovic Peyre
- Université Côte d'Azur, CNRS UMR 7284, Inserm U 1081, Institut de Recherche sur le Cancer et le Vieillissement de Nice (IRCAN), Centre Antoine Lacassagne, 06107, Nice, France
| | - Mickael Meyer
- Université Côte d'Azur, CNRS UMR 7284, Inserm U 1081, Institut de Recherche sur le Cancer et le Vieillissement de Nice (IRCAN), Centre Antoine Lacassagne, 06107, Nice, France
| | - Paul Hofman
- Université Côte d'Azur, CNRS UMR 7284, Inserm U 1081, Institut de Recherche sur le Cancer et le Vieillissement de Nice (IRCAN), Centre Antoine Lacassagne, 06107, Nice, France
| | - Jérémie Roux
- Université Côte d'Azur, CNRS UMR 7284, Inserm U 1081, Institut de Recherche sur le Cancer et le Vieillissement de Nice (IRCAN), Centre Antoine Lacassagne, 06107, Nice, France.
| |
Collapse
|
11
|
Cardoso Alves L, Corazza N, Micheau O, Krebs P. The multifaceted role of TRAIL signaling in cancer and immunity. FEBS J 2020; 288:5530-5554. [PMID: 33215853 DOI: 10.1111/febs.15637] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can lead to the induction of apoptosis in tumor or infected cells. However, activation of TRAIL signaling may also trigger nonapoptotic pathways in cancer and in nontransformed cells, that is, immune cells. Here, we review the current knowledge on noncanonical TRAIL signaling. The biological outcomes of TRAIL signaling in immune and malignant cells are presented and explained, with a focus on the role of TRAIL for natural killer (NK) cell function. Furthermore, we highlight the technical difficulties in dissecting the precise molecular mechanisms involved in the switch between apoptotic and nonapoptotic TRAIL signaling. Finally, we discuss the consequences thereof for a therapeutic manipulation of TRAIL in cancer and possible approaches to bypass these difficulties.
Collapse
Affiliation(s)
| | - Nadia Corazza
- Institute of Pathology, University of Bern, Switzerland
| | - Olivier Micheau
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | | |
Collapse
|
12
|
Wang JY, Lu PH, Lin WW, Wei YH, Chiu LY, Chern SR, Hung CF, Wu NL. Galectin-3 regulates UVB-induced inflammation in skin. J Dermatol Sci 2020; 98:119-127. [PMID: 32312639 DOI: 10.1016/j.jdermsci.2020.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Galectin-3 is widely expressed in many immunocytes and epithelial cells including skin keratinocytes. Galectin-3 can regulate immunological or inflammatory processes and plays a proinflammatory role in some disease models. Galectin-3 has a role in disorders related to ultraviolet (UV) photodamage such as apoptosis, skin squamous cell carcinoma and basal cell carcinoma. However, the evidence of galectin-3 in UVB-induced skin inflammation is still limited and the underlying molecular mechanism remains elusive. OBJECTIVE We aimed to investigate the effects of galectin-3 in human epidermal keratinocytes and in mice after UVB irradiation. METHODS Primary human epidermal keratinocytes with galectin-3 knockdown were used as the in vitro model. ELISA, QPCR, and western blotting were applied to evaluate the released cytokine, mRNA and protein expression. Histologic analysis, measurement of erythema and transepidermal water loss (TEWL) were applied to evaluate UVB-induced skin damage in galectin-3 knockout mice. RESULTS In UVB-irradiated human keratinocytes, galectin-3 knockdown downregulated the UVB-induced ASC crosslinking, cleavage of caspase-1, and formation of active IL-1β. Galectin-3 knockdown also decreased UVB-induced production of reactive oxygen species, p38 phosphorylation, and COX2 expression in human keratinocytes. After four days of UVB irradiation, galectin-3 knockout mice showed reduced gross erythema, histologic features of tissue inflammation, quantified levels of erythema and TEWL compared to wild type mice. The skin tissue lysate also showed less expression of active IL-1β and COX2 in galectin-3 knockout mice. CONCLUSION Galectin-3 may play a positive regulatory role in UVB-induced skin inflammation.
Collapse
Affiliation(s)
- Jen-Yu Wang
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan.
| | - Po-Hsuan Lu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yu-Hsuan Wei
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Ling-Ya Chiu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Schu-Rern Chern
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan.
| |
Collapse
|
13
|
El-Tahlawi S, Ezzat Mohammad N, Mohamed El-Amir A, Sayed Mohamed H. Survivin and insulin-like growth factor-I: potential role in the pathogenesis of acne and post-acne scar. Scars Burn Heal 2019; 5:2059513118818031. [PMID: 30675395 PMCID: PMC6330724 DOI: 10.1177/2059513118818031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDS/OBJECTIVES Acne is an inflammatory disease of the pilosebaceous unit (PSU). The over-expression of survivin and insulin-like growth factor (IGF)-I in some fibrotic disorders suggests a possible implication in the pathogenesis of acne and or post-acne scar. We aimed to evaluate their potential role in pathogenesis in acne and post-acne scar. METHODS Serum survivin and IGF-I levels were estimated in 30 patients with acne and post-acne scar compared to 30 controls. RESULTS There was a statistically significant difference in survivin and IGF-I levels between controls and patients (P < 0.05). However, there was no linear correlation between survivin and IGF-I. CONCLUSIONS Survivin and IGF-I could have a possible role in the pathogenesis of active acne and in post-inflammatory acne scar.
Collapse
|
14
|
Sajda T, Sinha AA. Autoantibody Signaling in Pemphigus Vulgaris: Development of an Integrated Model. Front Immunol 2018; 9:692. [PMID: 29755451 PMCID: PMC5932349 DOI: 10.3389/fimmu.2018.00692] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/21/2018] [Indexed: 01/10/2023] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin blistering disease effecting both cutaneous and mucosal epithelia. Blister formation in PV is known to result from the binding of autoantibodies (autoAbs) to keratinocyte antigens. The primary antigenic targets of pathogenic autoAbs are known to be desmoglein 3, and to a lesser extent, desmoglein 1, cadherin family proteins that partially comprise the desmosome, a protein structure responsible for maintaining cell adhesion, although additional autoAbs, whose role in blister formation is still unclear, are also known to be present in PV patients. Nevertheless, there remain large gaps in knowledge concerning the precise mechanisms through which autoAb binding induces blister formation. Consequently, the primary therapeutic interventions for PV focus on systemic immunosuppression, whose side effects represent a significant health risk to patients. In an effort to identify novel, disease-specific therapeutic targets, a multitude of studies attempting to elucidate the pathogenic mechanisms downstream of autoAb binding, have led to significant advancements in the understanding of autoAb-mediated blister formation. Despite this enhanced characterization of disease processes, a satisfactory explanation of autoAb-induced acantholysis still does not exist. Here, we carefully review the literature investigating the pathogenic disease mechanisms in PV and, taking into account the full scope of results from these studies, provide a novel, comprehensive theory of blister formation in PV.
Collapse
Affiliation(s)
- Thomas Sajda
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
15
|
Abstract
This review based on translational research predicts that the transcription factor p53 is the key effector of all anti-acne therapies. All-trans retinoic acid (ATRA) and isotretinoin (13-cis retinoic acid) enhance p53 expression. Tetracyclines and macrolides via inhibiting p450 enzymes attenuate ATRA degradation, thereby increase p53. Benzoyl peroxide and hydrogen peroxide elicit oxidative stress, which upregulates p53. Azelaic acid leads to mitochondrial damage associated with increased release of reactive oxygen species inducing p53. p53 inhibits the expression of androgen receptor and IGF-1 receptor, and induces the expression of IGF binding protein 3. p53 induces FoxO1, FoxO3, p21 and sestrin 1, sestrin 2, and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), the key inducer of isotretinoin-mediated sebocyte apoptosis explaining isotretinoin's sebum-suppressive effect. Anti-androgens attenuate the expression of miRNA-125b, a key negative regulator of p53. It can thus be concluded that all anti-acne therapies have a common mode of action, i.e., upregulation of the guardian of the genome p53. Immortalized p53-inactivated sebocyte cultures are unfortunate models for studying acne pathogenesis and treatment.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7a, 49076, Osnabrück, Germany.
| |
Collapse
|
16
|
Qian L, Xu F, Wang X, Jiang M, Wang J, Song W, Wu D, Shen Z, Feng D, Ling B, Cheng Y, Xiao W, Shan G, Zhou Y. LncRNA expression profile of ΔNp63α in cervical squamous cancers and its suppressive effects on LIF expression. Cytokine 2017; 96:114-122. [PMID: 28391028 DOI: 10.1016/j.cyto.2017.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/18/2017] [Accepted: 04/01/2017] [Indexed: 01/11/2023]
Abstract
We aim to determine the lncRNA targets of ΔNp63α in cervical cancer and molecular programs in cancerous differentiation. Different profiles of the lncRNAs were assayed and validated in overexpressing p63 SiHa cells (SiHa/ΔNp63α) and the control cell lines (SiHa/pCon). ENST00000422259, ENST00000447565 (Lnc-LIF-AS) and ENST00000469965, together with their related antisense mRNA DPYD (dihydropyrimidine dehydrogenase, a pyrimidine catabolic pathway gene), LIF (leukemia inhibitor factor) and FLNC (filamin C) were all notably differentially expressed in both ΔNp63α overexpression cells and knockdown cells. Here, we illustrated that ΔNp63α can inhibit the levels of LIF mRNA by direct transcription regulation and decrease LIF mRNA stability by suppressing the expression of Lnc-LIF-AS. An inverse interaction of LIF and ΔNp63α expression was as well validated in clinical samples of cervical cancer, and high level of LIF in cervical cancers was related with poor patient survival. The decrease of ΔNp63α also attenuated the differentiation of cervical cancerous cells. Suggesting that ΔNp63α may be form a complex network in regulation cervical cancerous differentiation.
Collapse
Affiliation(s)
- Lili Qian
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Fei Xu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Xiaolin Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, China
| | - Ming Jiang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Weiguo Song
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Dabao Wu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Dingqing Feng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Bin Ling
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yong Cheng
- Departments of Oncological Radiotherapy, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China
| | - Weihua Xiao
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, China.
| | - Ge Shan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, China.
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, China.
| |
Collapse
|
17
|
Melnik BC. The TRAIL to acne pathogenesis: let's focus on death pathways. Exp Dermatol 2016; 26:270-272. [PMID: 27541445 DOI: 10.1111/exd.13169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2016] [Indexed: 11/28/2022]
Abstract
This hypothesis presents acne as a PI3K-Akt-mTORC1-driven pro-survival disease of the sebaceous follicle with impaired TRAIL-mediated death signalling. It is predicted that anti-acne agents such as isotretinoin enhance death signalling and thereby readjust the disturbed balance of pro-survival and death signalling of the sebaceous follicle in acne vulgaris. For this purpose, immortalized sebocyte cultures are regarded as inapproproate models to study the key features of acne pathogenesis.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
18
|
Spleen Tyrosine Kinase Mediates EGFR Signaling to Regulate Keratinocyte Terminal Differentiation. J Invest Dermatol 2016; 136:192-201. [DOI: 10.1038/jid.2015.381] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 07/31/2015] [Accepted: 08/19/2015] [Indexed: 11/09/2022]
|
19
|
Kim SW, Park SY. Hypoxia‑mediated activation of autophagic flux inhibits apoptosis of keratinocytes via blocking tumor necrosis factor‑related apoptosis‑inducing ligand. Mol Med Rep 2015; 13:805-10. [PMID: 26648440 DOI: 10.3892/mmr.2015.4592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 06/30/2015] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑related apoptosis‑inducing ligand (TRAIL) is toxic against transformed tumor cells. Cornification is the terminal differentiation of keratinocytes and a specific form of programmed cell death caused by TRAIL that occurs in keratinocytes. Apoptosis can also be triggered when TRAIL induces expression of keratinocyte differentiation markers. The present study reported that hypoxia inhibits TRAIL‑induced apoptosis due to autophagic flux. It is well known that hypoxia activates autophagy in keratinocytes and reduces p62 protein levels. The present study demonstrated that hypoxia inhibited TRAIL‑mediated apoptosis and induced autophagic flux in HaCaT cells. In addition, autophagic flux‑inactivating reagents, including 3‑methyladenine and chloroquine, increased the TRAIL sensitivity of HaCaT cells exposed to hypoxia. In conclusion, these results indicated that inactivating autophagy increased TRAIL sensitivity in hypoxic HaCaT cells. Autophagy inhibitors may be beneficial in therapies using TRAIL against skin cancers.
Collapse
Affiliation(s)
- Sung-Wook Kim
- Department of Biochemistry, Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju, Jeonbuk 561‑756, Republic of Korea
| | - Sang-Youel Park
- Department of Biochemistry, Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju, Jeonbuk 561‑756, Republic of Korea
| |
Collapse
|
20
|
Wang JS, Wu D, Huang DY, Lin WW. TAK1 inhibition-induced RIP1-dependent apoptosis in murine macrophages relies on constitutive TNF-α signaling and ROS production. J Biomed Sci 2015; 22:76. [PMID: 26381601 PMCID: PMC4574455 DOI: 10.1186/s12929-015-0182-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/05/2015] [Indexed: 12/29/2022] Open
Abstract
Background Transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is a key regulator of signal cascades of TNF-α receptor and TLR4, and can induce NF-κB activation for preventing cell apoptosis and eliciting inflammation response. Results TAK1 inhibitor (TAKI) can decrease the cell viability of murine bone marrow-derived macrophages (BMDM), RAW264.7 and BV-2 cells, but not dermal microvascular endothelial cells, normal human epidermal keratinocytes, THP-1 monocytes, human retinal pigment epithelial cells, microglia CHME3 cells, and some cancer cell lines (CL1.0, HeLa and HCT116). In BMDM, TAKI-induced caspase activation and cell apoptosis were enhanced by lipopolysaccharide (LPS). Moreover, TAKI treatment increased the cytosolic and mitochondrial reactive oxygen species (ROS) production, and ROS scavengers NAC and BHA can inhibit cell death caused by TAKI. In addition, RIP1 inhibitor (necrostatin-1) can protect cells against TAKI-induced mitochondrial ROS production and cell apoptosis. We also observed the mitochondrial membrane potential loss after TAKI treatment and deterioration of oxygen consumption upon combination with LPS. Notably TNF-α neutralization antibody and inhibitor enbrel can decrease the cell death caused by TAKI. Conclusions TAKI-induced cytotoxicity is cell context specific, and apoptosis observed in macrophages is dependent on the constitutive autocrine action of TNF-α for RIP1 activation and ROS production.
Collapse
Affiliation(s)
- Jang-Shiun Wang
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan.,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Dean Wu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan. .,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
21
|
Flusberg DA, Sorger PK. Surviving apoptosis: life-death signaling in single cells. Trends Cell Biol 2015; 25:446-58. [PMID: 25920803 PMCID: PMC4570028 DOI: 10.1016/j.tcb.2015.03.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/19/2015] [Accepted: 03/19/2015] [Indexed: 12/16/2022]
Abstract
Tissue development and homeostasis are regulated by opposing pro-survival and pro-death signals. An interesting feature of the Tumor Necrosis Factor (TNF) family of ligands is that they simultaneously activate opposing signals within a single cell via the same ligand-receptor complex. The magnitude of pro-death events such as caspase activation and pro-survival events such as Nuclear Factor (NF)-κB activation vary not only from one cell type to the next but also among individual cells of the same type due to intrinsic and extrinsic noise. The molecules involved in these pro-survival and/or pro-death pathways, and the different phenotypes that result from their activities, have been recently reviewed. Here we focus on the impact of cell-to-cell variability in the strength of these opposing signals on shaping cell fate decisions.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Luyet C, Schulze K, Sayar BS, Howald D, Müller EJ, Galichet A. Preclinical studies identify non-apoptotic low-level caspase-3 as therapeutic target in pemphigus vulgaris. PLoS One 2015; 10:e0119809. [PMID: 25748204 PMCID: PMC4352034 DOI: 10.1371/journal.pone.0119809] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 02/03/2015] [Indexed: 02/07/2023] Open
Abstract
The majority of pemphigus vulgaris (PV) patients suffer from a live-threatening loss of intercellular adhesion between keratinocytes (acantholysis). The disease is caused by auto-antibodies that bind to desmosomal cadherins desmoglein (Dsg) 3 or Dsg3 and Dsg1 in mucous membranes and skin. A currently unresolved controversy in PV is whether apoptosis is involved in the pathogenic process. The objective of this study was to perform preclinical studies to investigate apoptotic pathway activation in PV pathogenesis with the goal to assess its potential for clinical therapy. For this purpose, we investigated mouse and human skin keratinocyte cultures treated with PV antibodies (the experimental Dsg3 monospecific antibody AK23 or PV patients IgG), PV mouse models (passive transfer of AK23 or PVIgG into adult and neonatal mice) as well as PV patients' biopsies (n=6). A combination of TUNEL assay, analyses of membrane integrity, early apoptotic markers such as cleaved poly-ADP-ribose polymerase (PARP) and the collapse of actin cytoskeleton failed to provide evidence for apoptosis in PV pathogenesis. However, the in vitro and in vivo PV models, allowing to monitor progression of lesion formation, revealed an early, transient and low-level caspase-3 activation. Pharmacological inhibition confirmed the functional implication of caspase-3 in major events in PV such as shedding of Dsg3, keratin retraction, proliferation including c-Myc induction, p38MAPK activation and acantholysis. Together, these data identify low-level caspase-3 activation downstream of disrupted Dsg3 trans- or cis-adhesion as a major event in PV pathogenesis that is non-synonymous with apoptosis and represents, unlike apoptotic components, a promising target for clinical therapy. At a broader level, these results posit that an impairment of adhesive functions in concert with low-level, non-lethal caspase-3 activation can evoke profound cellular changes which may be of relevance for other diseases including cancer.
Collapse
Affiliation(s)
- Camille Luyet
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Katja Schulze
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Beyza S. Sayar
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Denise Howald
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Eliane J. Müller
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Arnaud Galichet
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
23
|
Wu NL, Huang DY, Tsou HN, Lin YC, Lin WW. Syk Mediates IL−17-Induced CCL20 Expression by Targeting Act1-Dependent K63-Linked Ubiquitination of TRAF6. J Invest Dermatol 2015; 135:490-498. [DOI: 10.1038/jid.2014.383] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 12/21/2022]
|
24
|
Lin YC, Huang DY, Wang JS, Lin YL, Hsieh SL, Huang KC, Lin WW. Syk is involved in NLRP3 inflammasome-mediated caspase-1 activation through adaptor ASC phosphorylation and enhanced oligomerization. J Leukoc Biol 2015; 97:825-835. [PMID: 25605870 DOI: 10.1189/jlb.3hi0814-371rr] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/03/2014] [Accepted: 11/30/2014] [Indexed: 12/27/2022] Open
Abstract
NLRP3 is the most crucial member of the NLR family, as it detects the existence of pathogen invasion and self-derived molecules associated with cellular damage. Several studies have reported that excessive NLRP3 inflammasome-mediated caspase-1 activation is a key factor in the development of diseases. Recent studies have reported that Syk is involved in pathogen-induced NLRP3 inflammasome activation; however, the detailed mechanism linking Syk to NLRP3 inflammasome remains unclear. In this study, we showed that Syk mediates NLRP3 stimuli-induced processing of procaspase-1 and the consequent activation of caspase-1. Moreover, the kinase activity of Syk is required to potentiate caspase-1 activation in a reconstituted NLRP3 inflammasome system in HEK293T cells. The adaptor protein ASC bridges NLRP3 with the effector protein caspase-1. Herein, we find that Syk can associate directly with ASC and NLRP3 by its kinase domain but interact indirectly with procaspase-1. Syk can phosphorylate ASC at Y146 and Y187 residues, and the phosphorylation of both residues is critical to enhance ASC oligomerization and the recruitment of procaspase-1. Together, our results reveal a new molecular pathway through which Syk promotes NLRP3 inflammasome formation, resulting from the phosphorylation of ASC. Thus, the control of Syk activity might be effective to modulate NLRP3 inflammasome activation and treat NLRP3-related immune diseases.
Collapse
Affiliation(s)
- Ying-Cing Lin
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Duen-Yi Huang
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jang-Shiun Wang
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ling Lin
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shie-Liang Hsieh
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Chin Huang
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Wan Lin
- *Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences and Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan; and Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
25
|
Turunen A, Hukkanen V, Nygårdas M, Kulmala J, Syrjänen S. The combined effects of irradiation and herpes simplex virus type 1 infection on an immortal gingival cell line. Virol J 2014; 11:125. [PMID: 25005804 PMCID: PMC4105526 DOI: 10.1186/1743-422x-11-125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/03/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Oral mucosa is frequently exposed to Herpes simplex virus type 1 (HSV-1) infection and irradiation due to dental radiography. During radiotherapy for oral cancer, the surrounding clinically normal tissues are also irradiated. This prompted us to study the effects of HSV-1 infection and irradiation on viability and apoptosis of oral epithelial cells. METHODS Immortal gingival keratinocyte (HMK) cells were infected with HSV-1 at a low multiplicity of infection (MOI) and irradiated with 2 Gy 24 hours post infection. The cells were then harvested at 24, 72 and 144 hours post irradiation for viability assays and qRT-PCR analyses for the apoptosis-related genes caspases 3, 8, and 9, bcl-2, NFκB1, and viral gene VP16. Mann-Whitney U-test was used for statistical calculations. RESULTS Irradiation improved the cell viability at 144 hours post irradiation (P = 0.05), which was further improved by HSV-1 infection at MOI of 0.00001 (P = 0.05). Simultaneously, the combined effects of infection at MOI of 0.0001 and irradiation resulted in upregulation in NFκB1 (P = 0.05). The combined effects of irradiation and HSV infection also significantly downregulated the expression of caspases 3, 8, and 9 at 144 hours (P = 0.05) whereas caspase 3 and 8 significantly upregulated in non-irradiated, HSV-infected cells as compared to uninfected controls (P = 0.05). Infection with 0.0001 MOI downregulated bcl-2 in non-irradiated cells but was upregulated by 27% after irradiation when compared to non-irradiated infected cells (P = 0.05). Irradiation had no effect on HSV-1 shedding or HSV gene expression at 144 hours. CONCLUSIONS HSV-1 infection may improve the viability of immortal cells after irradiation. The effect might be related to inhibition of apoptosis.
Collapse
Affiliation(s)
- Aaro Turunen
- Institute of Dentistry, Department of Oral Pathology, University of Turku, Lemminkäisenkatu 2, 20520 Turku, Finland.
| | | | | | | | | |
Collapse
|
26
|
Huang CT, Huang DY, Hu CJ, Wu D, Lin WW. Energy adaptive response during parthanatos is enhanced by PD98059 and involves mitochondrial function but not autophagy induction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:531-43. [PMID: 24321770 DOI: 10.1016/j.bbamcr.2013.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 02/08/2023]
Abstract
Parthanatos is a programmed necrotic demise characteristic of ATP (adenosine triphosphate) consumption due to NAD+ (nicotinamide adenine dinucleotide) depletion by poly(ADP-ribose) polymerase 1 (PARP1)-dependent poly(ADP-ribosyl)ation on target proteins. However, how the bioenergetics is adaptively regulated during parthanatos, especially under the condition of macroautophagy deficiency, remains poorly characterized. Here, we demonstrated that the parthanatic inducer N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) triggered ATP depletion followed by recovery in mouse embryonic fibroblasts (MEFs). Notably, Atg5-/- MEFs showed great susceptibility to MNNG with disabled ATP-producing capacity. Moreover, the differential energy-adaptive responses in wild-type (WT) and Atg5-/- MEFs were unequivocally worsened by inhibition ofAMP-activated protein kinase (AMPK), sirtuin 1 (SIRT1), and mitochondrial activity. Importantly, Atg5-/- MEFs disclosed diminished SIRT1 and mitochondrial activity essential to the energy restoration during parthanatos. Strikingly, however, parthanatos cannot be exasperated by bafilomycin A1 and MNNG neither provokes microtubule-associated protein 1A/1B-light chain 3 (LC3) lipidation and p62 elimination, suggesting that parthanatos does not induce autophagic flux. Intriguingly, we reported unexpectedly that PD98059, even at low concentration insufficient to inhibit MEK, can promote mitochondrial activity and facilitate energy-restoring process during parthanatos, without modulating DNA damage responses as evidenced by PARP1 activity, p53 expression, and gammaH2AX (H2A histone family, member X (H2AX), phosphorylated on Serine 139) induction. Therefore, we propose that Atg5 deficiency confers an infirmity to overcome the energy crisis during parthanatos and further underscore the deficits in mitochondrial quality control, but not incapability of autophagy induction, that explain the vulnerability in Atg5-deficient cells. Collectively, our results provide a comprehensive energy perspective for an improved treatment to alleviate parthanatos-related tissue necrosis and disease progression and also provide a future direction for drug development on the basis of PD98059 as an efficacious compound against parthanatos.
Collapse
|
27
|
Fischer B, Metzger M, Richardson R, Knyphausen P, Ramezani T, Franzen R, Schmelzer E, Bloch W, Carney TJ, Hammerschmidt M. p53 and TAp63 promote keratinocyte proliferation and differentiation in breeding tubercles of the zebrafish. PLoS Genet 2014; 10:e1004048. [PMID: 24415949 PMCID: PMC3886889 DOI: 10.1371/journal.pgen.1004048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 11/04/2013] [Indexed: 11/18/2022] Open
Abstract
p63 is a multi-isoform member of the p53 family of transcription factors. There is compelling genetic evidence that ΔNp63 isoforms are needed for keratinocyte proliferation and stemness in the developing vertebrate epidermis. However, the role of TAp63 isoforms is not fully understood, and TAp63 knockout mice display normal epidermal development. Here, we show that zebrafish mutants specifically lacking TAp63 isoforms, or p53, display compromised development of breeding tubercles, epidermal appendages which according to our analyses display more advanced stratification and keratinization than regular epidermis, including continuous desquamation and renewal of superficial cells by derivatives of basal keratinocytes. Defects are further enhanced in TAp63/p53 double mutants, pointing to partially redundant roles of the two related factors. Molecular analyses, treatments with chemical inhibitors and epistasis studies further reveal the existence of a linear TAp63/p53->Notch->caspase 3 pathway required both for enhanced proliferation of keratinocytes at the base of the tubercles and their subsequent differentiation in upper layers. Together, these studies identify the zebrafish breeding tubercles as specific epidermal structures sharing crucial features with the cornified mammalian epidermis. In addition, they unravel essential roles of TAp63 and p53 to promote both keratinocyte proliferation and their terminal differentiation by promoting Notch signalling and caspase 3 activity, ensuring formation and proper homeostasis of this self-renewing stratified epithelium. The mammalian epidermis is a stratified self-renewing epithelium, in which cell loss at the surface is properly balanced by cell proliferation in basal layers to ensure tissue homeostasis. But how is this balance genetically controlled? Here, we address this question in zebrafish breeding tubercles, epidermal appendages in which keratinocytes undergo more advanced differentiation processes than in regular fish epidermis, sharing crucial features with the cornified mammalian skin. We identify a linear pathway consisting of the transcription factor p53 and its close relative TAp63, which activate Notch signalling and thereby caspase 3 to promote terminal differentiation and eventual shedding of keratinocytes in upper tubercle layers, while at the same time employing non-cell autonomous mechanisms to promote keratinocyte proliferation at the tubercle base, thereby ensuring proper development and homeostasis of this self-renewing tissue. Such a two-fold function of the pathway is consistent with the formerly reported dual role of a caspase during wing regeneration in the fruitfly. Our findings will help to better understand the seemingly contrary effects described for TAp63 in different mammalian systems, while demonstrating partial functional redundancy between p53 and TAp63 during epidermal development in fish.
Collapse
Affiliation(s)
- Boris Fischer
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Manuel Metzger
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Rebecca Richardson
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Philipp Knyphausen
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Thomas Ramezani
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Rainer Franzen
- Cell Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Elmon Schmelzer
- Cell Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | | | - Matthias Hammerschmidt
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
28
|
Wu NL, Huang DY, Hsieh SL, Hsiao CH, Lee TA, Lin WW. EGFR-driven up-regulation of decoy receptor 3 in keratinocytes contributes to the pathogenesis of psoriasis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1538-48. [PMID: 23707413 DOI: 10.1016/j.bbadis.2013.05.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/13/2013] [Accepted: 05/16/2013] [Indexed: 02/07/2023]
Abstract
Decoy receptor 3 (DcR3) is a soluble receptor of Fas ligand (FasL), LIGHT (TNFSF14) and TNF-like molecule 1A (TL1A) and plays pleiotropic roles in many inflammatory and autoimmune disorders and malignant diseases. In cutaneous biology, DcR3 is expressed in primary human epidermal keratinocytes and is upregulated in skin lesions in psoriasis, which is characterized by chronic inflammation and angiogenesis. However, the regulatory mechanisms of DcR3 over-expression in skin lesions of psoriasis are unknown. Here, we demonstrate that DcR3 can be detected in both dermal blood vessels and epidermal layers of psoriatic skin lesions. Analysis of serum samples showed that DcR3 was elevated, but FasL was downregulated in psoriatic patients compared with normal individuals. Additional cell studies revealed a central role of epidermal growth factor receptor (EGFR) in controlling the basal expression of DcR3 in keratinocytes. Activation of EGFR by epidermal growth factor (EGF) and transforming growth factor (TGF)-α strikingly upregulated DcR3 production. TNF-αenhanced DcR3 expression in both keratinocytes and endothelial cells compared with various inflammatory cytokines involved in psoriasis. Additionally, TNF-α-enhanced DcR3 expression in keratinocytes was inhibited when EGFR was knocked down or EGFR inhibitor was used. The NF-κB pathway was critically involved in the molecular mechanisms underlying the action of EGFR and inflammatory cytokines. Collectively, the novel regulatory mechanisms of DcR3 expression in psoriasis, particularly in keratinocytes and endothelial cells, provides new insight into the pathogenesis of psoriasis and may also contribute to the understanding of other diseases that involve DcR3 overexpression.
Collapse
Affiliation(s)
- Nan-Lin Wu
- Department of Pharmacology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Non-canonical kinase signaling by the death ligand TRAIL in cancer cells: discord in the death receptor family. Cell Death Differ 2013; 20:858-68. [PMID: 23579241 DOI: 10.1038/cdd.2013.28] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based therapy is currently evaluated in clinical studies as a tumor cell selective pro-apoptotic approach. However, besides activating canonical caspase-dependent apoptosis by binding to TRAIL-specific death receptors, the TRAIL ligand can activate non-canonical cell survival or proliferation pathways in resistant tumor cells through the same death receptors, which is counterproductive for therapy. Even more, recent studies indicate metastases-promoting activity of TRAIL. In this review, the remarkable dichotomy in TRAIL signaling is highlighted. An overview of the currently known mechanisms involved in non-canonical TRAIL signaling and the subsequent activation of various kinases is provided. These kinases include RIP1, IκB/ NF-κB, MAPK p38, JNK, ERK1/2, MAP3K TAK1, PKC, PI3K/Akt and Src. The functional consequences of their activation, often being stimulation of tumor cell survival and in some cases enhancement of their invasive behavior, are discussed. Interestingly, the non-canonical responses triggered by TRAIL in resistant tumor cells resemble that of TRAIL-induced signals in non-transformed cells. Better knowledge of the mechanism underlying the dichotomy in TRAIL receptor signaling may provide markers for selecting patients who will likely benefit from TRAIL-based therapy and could provide a rationalized basis for combination therapies with TRAIL death receptor-targeting drugs.
Collapse
|
30
|
Brocklehurst K, Philpott MP. Cysteine proteases: mode of action and role in epidermal differentiation. Cell Tissue Res 2013; 351:237-44. [PMID: 23344364 DOI: 10.1007/s00441-013-1557-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/03/2013] [Indexed: 12/31/2022]
Abstract
Desquamation or cell shedding in mammalian skin is known to involve serine proteases, aspartic proteases and glycosidases. In addition, evidence continues to accumulate that papain-like cysteine proteases and an inhibitor cystatin M/E largely confined to the cutaneous epithelia also play key roles in the process. This involves the complete proteolysis of cell adhesive structures of the stratum corneum, the corneodesmosomes and notably of the desmogleins. Continual cell replacement in the epidermis is the result of the balance between the loss of the outer squames and mitosis of the cells in the basal cell layer. This article provides a brief account of the salient features of the characteristics and catalytic mechanism of cysteine proteases, followed by a discussion of the relevant epidermal biology. The proteases include the asparaginyl endopeptidase legumain, which exerts a strict specificity for the hydrolysis of asparaginyl bonds, cathepsin-V and cathepsin-L. The control of these enzymes by cystatin M/E regulates the processing of transglutaminases and is crucial in the biochemical pathway responsible for regulating the cross-linking and desquamation of the stratum corneum. In addition, caspase-14 has now been shown to play a major part in epidermal maturation. Uncontrolled proteolytic activity leads to abnormal hair follicle formation and deleterious effects on the skin barrier function.
Collapse
Affiliation(s)
- Keith Brocklehurst
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | | |
Collapse
|
31
|
Abstract
Dermatitis herpetiformis (DH) is a gluten-sensitive autoimmune blistering disorder with a chronic-relapsing course. Very recently, several Authors reported atypical cases of patients with DH, suggesting that different clinical subsets may exist at least among different ethnicities and that the classical picture of DH probably need a significant revision. Moreover, different pathogenetic aspects of the disease are currently under investigation, including the role of epidermal transglutaminase, apoptosis and inflammatory cells in the occurrence of skin lesions, in order to explain why only a subgroup of celiac patients will develop DH. Finally, although gluten-free diet is still regarded as the only curative approach to the disease, it is very hard to comply with and even small amounts of gluten can re-activate the disease. Therefore, different therapeutical approaches for the spectrum DH/celiac disease are still under investigation. In the present paper, the most recent advances in DH will be discussed, and a novel interpretation of the disease based on the data emerging from the Literature will be proposed.
Collapse
|
32
|
The expression of selected proapoptotic molecules in dermatitis herpetiformis. Clin Dev Immunol 2012; 2012:178340. [PMID: 22778762 PMCID: PMC3388338 DOI: 10.1155/2012/178340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 04/06/2012] [Accepted: 04/25/2012] [Indexed: 11/17/2022]
Abstract
The role of the process of apoptosis is investigated in the pathogenesis of many autoimmune diseases; however at present, there is not much information about its role in dermatitis herpetiformis. Skin biopsies were taken from 18 DH patients and from 10 healthy subjects. The localization and expression of Bax, Fas, FasL, TRAIL, TRAIL-R in skin lesions, and perilesional skin were studied by immunohistochemistry. Expression of Bax, Fas, and Fas ligand was detected in the keratinocytes in skin biopsies from DH patients. Expression of TRAIL and TRAIL receptor was confirmed in epidermis, infiltration cells, and some fibroblasts. The expression of examined molecules in biopsies from healthy people was observed only in single cells. There were statistically significant differences between lesional, perilesional, and healthy skin of control group in Bax expression analysis and between lesional skin and control group in Fas, FasL, and TRAIL expression. There were statistically significant differences between control group and perilesional skin in Bax and FasL expression. Our results show that selected proapoptotic molecules may take part in pathogenesis of dermatitis herpetiformis, but the role of apoptosis in this process is not clear.
Collapse
|
33
|
Rethi B, Eidsmo L. FasL and TRAIL signaling in the skin during cutaneous leishmaniasis - implications for tissue immunopathology and infectious control. Front Immunol 2012; 3:163. [PMID: 22723798 PMCID: PMC3377931 DOI: 10.3389/fimmu.2012.00163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 05/31/2012] [Indexed: 01/09/2023] Open
Abstract
Cutaneous leishmaniasis (CL) is associated with chronic inflammation and ulceration of the skin. Tissue macrophages serve as host cells and immune activation is necessary for parasite clearance. The balance between immune-mediated tissue destruction and successful clearance of infection is delicate and ulceration has been proposed to be a result of infiltration of activated immune cells into the skin. FasL and TRAIL play a dual role in skin homeostasis through induction of apoptosis as well as proinflammatory signaling. During leishmaniasis, dysregulation of both FasL and TRAIL has been described by us and others but the resulting pathogenic effects in the skin during human leishmaniasis are not fully elucidated. Targeting disease specific immune deviations has proven to be a promising new approach for the therapy of autoimmune diseases. Potentially, targeting FasL or TRAIL in combination with microcidals could offer a future treatment strategy to reduce the disfiguring immunopathology associated with CL. In this mini review we will discuss how FasL and TRAIL-induced signaling may influence on the extent of tissue inflammation and the efficacy of parasite clearance in leishmaniasis.
Collapse
Affiliation(s)
- Bence Rethi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
34
|
Shirley S, Morizot A, Micheau O. Regulating TRAIL receptor-induced cell death at the membrane : a deadly discussion. Recent Pat Anticancer Drug Discov 2011; 6:311-23. [PMID: 21756247 PMCID: PMC3204462 DOI: 10.2174/157489211796957757] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 02/20/2011] [Accepted: 02/20/2011] [Indexed: 12/20/2022]
Abstract
The use of TRAIL/APO2L and monoclonal antibodies targeting TRAIL receptors for cancer therapy holds great promise, due to their ability to restore cancer cell sensitivity to apoptosis in association with conventional chemotherapeutic drugs in a large variety of tumors. TRAIL-induced cell death is tightly regulated right from the membrane and at the DISC (Death-Inducing Signaling Complex) level. The following patent and literature review aims to present and highlight recent findings of the deadly discussion that determines tumor cell fate upon TRAIL engagement.
Collapse
Affiliation(s)
- Sarah Shirley
- INSERM, U866, Dijon, F-21079 France; Faculty of Medicine and Pharmacy, University of Bourgogne, Dijon, F-21079 France.
| | | | | |
Collapse
|