1
|
Cho YH, Park JE. Anti-Inflammatory and Autophagy Activation Effects of 7-Methylsulfonylheptyl Isothiocyanate Could Suppress Skin Aging: In Vitro Evidence. Antioxidants (Basel) 2024; 13:1282. [PMID: 39594424 PMCID: PMC11591029 DOI: 10.3390/antiox13111282] [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: 09/11/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
Skin inflammation, characterized by redness, swelling, and discomfort, is exacerbated by oxidative stress, where compounds like 7-methylsulfonylheptyl isothiocyanate (7-MSI) from cruciferous plants exhibit promising antioxidant and anti-inflammatory properties, though their effects on skin aging and underlying mechanisms involving the NLRP3 inflammasome and autophagy are not fully elucidated. NLRP3 is a crucial inflammasome involved in regulating inflammatory responses, and our study addresses its activation and associated physiological effects. Using biochemical assays such as ELISA, RT-qPCR, Western blotting, confocal microscopy, and RNA interference, we evaluated 7-MSI's impact on cytokine production, protein expression, and genetic regulation in Raw 264.7 and RAW-ASC cells. 7-MSI significantly reduced TNF-α, IL-1β, IL-6, COX-2, and PGE transcription levels in LPS-stimulated Raw 264.7 cells, indicating potent anti-inflammatory effects. It also inhibited NF-κB signaling and NLRP3 inflammasome activity, demonstrating its role in preventing the nuclear translocation of NF-κB and reducing caspase-1 and IL-1β production. In terms of autophagy, 7-MSI enhanced the expression of Beclin-1, LC3, and Atg12 while reducing phospho-mTOR levels, suggesting an activation of autophagy. Moreover, it effectively decreased ROS production induced by LPS. The interaction between autophagy and inflammasome regulation was further confirmed through experiments showing that interference with autophagy-related genes altered the effects of 7-MSI on cytokine production. Collectively, this study demonstrates that 7-MSI promotes autophagy, including ROS removal, and to suppress inflammation, we suggest the potential use of 7-MSI as a skin care and disease treatment agent.
Collapse
Affiliation(s)
- Yeong Hee Cho
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Republic of Korea;
- Department of Agricultural Biology, National Institute of Agricultural Sciences, RDA, Wanju-gun 55365, Republic of Korea
| | - Jung Eun Park
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Republic of Korea;
- BK21-Four Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
2
|
Kuncorojakti S, Pratama AZA, Antujala CA, Harijanto CTB, Arsy RK, Kurniawan PA, Tjahjono Y, Hendriati L, Widodo T, Aswin A, Diyantoro D, Wijaya AY, Rodprasert W, Susilowati H. Acceleration of wound healing using adipose mesenchymal stem cell secretome hydrogel on partial-thickness cutaneous thermal burn wounds: An in vivo study in rats. Vet World 2024; 17:1545-1554. [PMID: 39185045 PMCID: PMC11344119 DOI: 10.14202/vetworld.2024.1545-1554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/21/2024] [Indexed: 08/27/2024] Open
Abstract
Background and Aim The intricate healing process involves distinct sequential and overlapping phases in thermal injury. To maintain the zone of stasis in Jackson's burn wound model, proper wound intervention is essential. The extent of research on the histoarchitecture of thermal wound healing and the application of mesenchymal stem cell (MSC)-free-based therapy is limited. This study aimed to assess the efficacy of MSC-secretome-based hydrogel for treating partial-thickness cutaneous thermal burn wounds. Materials and Methods Eighteen male Wistar rats were divided into three groups, namely the hydrogel base (10 mg), hydrogel secretome (10 mg) and Bioplacenton™ (10 mg) treatment groups. All groups were treated twice a day (morning and evening) for 7 days. Skin tissue samples from the animals were processed for histological evaluation using the formalin-fixed paraffin-embedded method on days 3 and 7. Results This study's findings showed that secretome hydrogel expedited thermal burn wound healing, decreasing residual burn area, boosting collagen deposition and angiogenesis, guiding scar formation, and influencing the inflammation response facilitated by polymorphonuclear leukocytes and macrophages. Conclusion The secretome hydrogel significantly improves healing outcomes in partial-thickness cutaneous thermal burn wounds. The administration of secretome hydrogel accelerates the reduction of the residual burn area and promotes fibroblast proliferation and collagen density. The repairment of histo-architecture of the damaged tissue was also observed such as the reduction of burn depth, increased angiogenesis and epidermal scar index while the decreased dermal scar index. Furthermore, the secretome hydrogel can modulate the immunocompetent cells by decreasing the polymorphonuclear and increasing the mononuclear cells. Thus, it effectively and safely substitutes for thermal injury stem cell-free therapeutic approaches. The study focuses on the microscopical evaluation of secretome hydrogel; further research to investigate at the molecular level may be useful in predicting the beneficial effect of secretome hydrogel in accelerating wound healing.
Collapse
Affiliation(s)
- Suryo Kuncorojakti
- Division of Veterinary Anatomy, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
- Research Centre for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | | | - Cahya Asri Antujala
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | | | - Rozak Kurnia Arsy
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Putut Andika Kurniawan
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Yudy Tjahjono
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Lucia Hendriati
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Teguh Widodo
- Department of Pharmaceutics Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Ahmad Aswin
- Research Centre for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Diyantoro Diyantoro
- Research Centre for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Health, Faculty of Vocational Studies, Universitas Airlangga, Surabaya, Indonesia
| | - Andi Yasmin Wijaya
- Research Centre for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Helen Susilowati
- Research Centre for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
3
|
Minoretti P. Chronic Pruritus Alleviation in the Elderly Through Drug-Free Autophagy Activation by Magnetized Saline Water: A Case Series. Cureus 2024; 16:e64428. [PMID: 39130837 PMCID: PMC11317106 DOI: 10.7759/cureus.64428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Chronic pruritus is a common and distressing condition in the elderly population, frequently associated with various underlying systemic diseases and age-related skin changes. Conventional treatments, such as emollients and moisturizers, may not invariably provide adequate relief. Magnetized saline water has previously been shown to activate autophagy, a cellular process involved in maintaining skin barrier function, reducing inflammaging, and modulating neuropathic pain. This case series investigated the efficacy of a topical serum containing magnetized saline water in managing chronic pruritus with diverse etiologies in elderly patients. Five patients aged 69-80 years, presenting with chronic pruritus lasting two to six months, were instructed to apply the serum daily to the most affected areas for a minimum of 14 consecutive days. Pruritus severity was assessed using the 12-Item Pruritus Severity Scale (12-PSS) at baseline and post-intervention. The underlying causes of pruritus included end-stage renal disease, type 2 diabetes mellitus with peripheral neuropathy, advanced liver fibrosis, and xerosis cutis. All five patients reported a substantial improvement in pruritus severity following the application of the magnetized saline water serum, with post-intervention 12-PSS scores decreasing by 3-5 points. The serum was well-tolerated, and no adverse effects were reported. These findings suggest that topical formulations containing magnetized saline water may be a promising alternative or adjunctive therapy for managing chronic pruritus in the elderly population. However, clinical trials are needed to confirm these findings, elucidate the precise mechanisms of action, and establish optimal treatment protocols.
Collapse
|
4
|
Deeudomwongsa P, Chumsaengsri C, Aristizabal M, Kiatsurayanon C. Exploring the potential of intradermal platelet-rich plasma in treating acquired bilateral nevus of Ota-like macule (Hori's nevus): A pilot study. J Cosmet Dermatol 2024; 23:803-811. [PMID: 38088245 DOI: 10.1111/jocd.16135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND Hori's nevus is a common and challenging dermatological condition, often complicated by post-inflammatory hyperpigmentation following treatment. Platelet-rich plasma (PRP) has demonstrated efficacy in the treatment of hyperpigmentation disorders such as melasma and periorbital darkening. Given the benefits and minimally invasive nature of PRP treatments, exploring its application in managing Hori's nevus through further investigation is worthwhile. AIMS To evaluate the safety and effectiveness of intradermal PRP therapy for the treatment of Hori's nevus. METHODS Ten female patients received bilateral intradermal PRP injections every 2 weeks for a total of four treatments. The modified dermal pigmentation and severity index (mDPASI), mean melanin index (MI), brightening score, patient self-assessment, and clinical photographs were evaluated at 2, 4, 8, and 12 weeks post-treatment. Adverse events were also recorded to determine treatment safety. RESULTS At 12 weeks post-treatment, mDPASI decreased 38.86%, from 0.929 ± 0.617 to 0.568 ± 0.415 (p < 0.05). The mean melanin index decreased 12.75%, from 208.650 ± 26.319 to 182.052 ± 17.028 (p < 0.05). In addition, the mean brightness score evaluated by two experts was 1.4, indicating 25-50% improvement. At the end of the study, 50% of the patients reported 50-75% improvement. Side effects included pain, mild edema, and bruising, which resolved spontaneously within 3 days. No serious side effects were found. CONCLUSION Our results suggest that intradermal PRP therapy may be a safe and effective alternative for the treatment of Hori's nevus and can complement conventional interventions. However, further research with a larger sample size, control groups, and longer follow-up is needed to confirm these findings.
Collapse
Affiliation(s)
- Pasinee Deeudomwongsa
- College of Medicine, Rangsit University, Bangkok, Thailand
- Dermatology, Institute of Dermatology, Bangkok, Thailand
| | | | - Miguel Aristizabal
- ADEI - Aesthetics & Dermatology Institute, Bogota, Colombia
- Mayo Clinic College of Medicine, Jacksonville, Florida, USA
| | | |
Collapse
|
5
|
Kuczyńska M, Moskot M, Gabig-Cimińska M. Insights into Autophagic Machinery and Lysosomal Function in Cells Involved in the Psoriatic Immune-Mediated Inflammatory Cascade. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0005. [PMID: 38409665 DOI: 10.2478/aite-2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/08/2023] [Indexed: 02/28/2024]
Abstract
Impaired autophagy, due to the dysfunction of lysosomal organelles, contributes to maladaptive responses by pathways central to the immune system. Deciphering the immune-inflammatory ecosystem is essential, but remains a major challenge in terms of understanding the mechanisms responsible for autoimmune diseases. Accumulating evidence implicates a role that is played by a dysfunctional autophagy-lysosomal pathway (ALP) and an immune niche in psoriasis (Ps), one of the most common chronic skin diseases, characterized by the co-existence of autoimmune and autoinflammatory responses. The dysregulated autophagy associated with the defective lysosomal system is only one aspect of Ps pathogenesis. It probably cannot fully explain the pathomechanism involved in Ps, but it is likely important and should be seriously considered in Ps research. This review provides a recent update on discoveries in the field. Also, it sheds light on how the dysregulation of intracellular pathways, coming from modulated autophagy and endolysosomal trafficking, characteristic of key players of the disease, i.e., skin-resident cells, as well as circulating immune cells, may be responsible for immune impairment and the development of Ps.
Collapse
Affiliation(s)
- Martyna Kuczyńska
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | - Marta Moskot
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
6
|
Dernovics Á, Seprényi G, Rázga Z, Ayaydin F, Veréb Z, Megyeri K. Phenol-Soluble Modulin α3 Stimulates Autophagy in HaCaT Keratinocytes. Biomedicines 2023; 11:3018. [PMID: 38002017 PMCID: PMC10669503 DOI: 10.3390/biomedicines11113018] [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: 09/02/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Phenol-soluble modulins (PSMs) are pore-forming toxins (PFTs) produced by staphylococci. PSMs exert diverse cellular effects, including lytic, pro-apoptotic, pro-inflammatory and antimicrobial actions. Since the effects of PSMs on autophagy have not yet been reported, we evaluated the autophagic activity in HaCaT keratinocytes treated with recombinant PSMα3. METHODS The autophagic flux and levels of autophagic marker proteins were determined using Western blot analysis. Subcellular localization of LC3B and Beclin-1 was investigated using an indirect immunofluorescence assay. The ultrastructural features of control and PSMα3-treated cells were evaluated via transmission electron microscopy. Cytoplasmic acidification was measured via acridine orange staining. Phosphorylation levels of protein kinases, implicated in autophagy regulation, were studied using a phospho-kinase array and Western blot analysis. RESULTS PSMα3 facilitated the intracellular redistribution of LC3B, increased the average number of autophagosomes per cell, promoted the development of acidic vesicular organelles, elevated the levels of LC3B-II, stimulated autophagic flux and triggered a significant decrease in the net autophagic turnover rate. PSMα3 induced the accumulation of autophagosomes/autolysosomes, amphisomes and multilamellar bodies at the 0.5, 6 and 24 h time points, respectively. The phospho-Akt1/2/3 (T308 and S473), and phospho-mTOR (S2448) levels were decreased, whereas the phospho-Erk1/2 (T202/Y204 and T185/Y187) level was increased in PSMα3-treated cells. CONCLUSIONS In HaCaT keratinocytes, PSMα3 stimulates autophagy. The increased autophagic activity elicited by sub-lytic PSM concentrations might be an integral part of the cellular defense mechanisms protecting skin homeostasis.
Collapse
Affiliation(s)
- Áron Dernovics
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| | - György Seprényi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Kossuth L. sgt. 40., H-6724 Szeged, Hungary;
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Állomás u. 2, H-6720 Szeged, Hungary;
| | - Ferhan Ayaydin
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM) Nonprofit Ltd., Római krt. 21., H-6723 Szeged, Hungary;
- Laboratory of Cellular Imaging, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62., H-6726 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Korányi Fasor 6, H-6720 Szeged, Hungary;
- Biobank, University of Szeged, H-6720 Szeged, Hungary
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Klára Megyeri
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| |
Collapse
|
7
|
Hunt M, Torres M, Bachar-Wikström E, Wikström JD. Multifaceted roles of mitochondria in wound healing and chronic wound pathogenesis. Front Cell Dev Biol 2023; 11:1252318. [PMID: 37771375 PMCID: PMC10523588 DOI: 10.3389/fcell.2023.1252318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Mitochondria are intracellular organelles that play a critical role in numerous cellular processes including the regulation of metabolism, cellular stress response, and cell fate. Mitochondria themselves are subject to well-orchestrated regulation in order to maintain organelle and cellular homeostasis. Wound healing is a multifactorial process that involves the stringent regulation of several cell types and cellular processes. In the event of dysregulated wound healing, hard-to-heal chronic wounds form and can place a significant burden on healthcare systems. Importantly, treatment options remain limited owing to the multifactorial nature of chronic wound pathogenesis. One area that has received more attention in recent years is the role of mitochondria in wound healing. With regards to this, current literature has demonstrated an important role for mitochondria in several areas of wound healing and chronic wound pathogenesis including metabolism, apoptosis, and redox signalling. Additionally, the influence of mitochondrial dynamics and mitophagy has also been investigated. However, few studies have utilised patient tissue when studying mitochondria in wound healing, instead using various animal models. In this review we dissect the current knowledge of the role of mitochondria in wound healing and discuss how future research can potentially aid in the progression of wound healing research.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Zhang Q, Zhang Q, Yan X, Wang L, Yuan X. Wrinkled topography regulates osteogenesis via autophagy-mediated Wnt/β-catenin signaling pathway in MC3T3-E1 cells. Arch Oral Biol 2023; 151:105700. [PMID: 37094411 DOI: 10.1016/j.archoralbio.2023.105700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/30/2023] [Accepted: 04/13/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE In this study, we aimed to evaluate the effects of different dimensional wrinkled in topography on the osteogenic differentiation of MC3T3-E1 cells and explored the underlying mechanisms. DESIGN Polydimethylsiloxane (PDMS) with a wrinkled topography was synthesized using an elastomer base and crosslinking while observing by atomic force microscopy. MC3T3-E1 proliferation was detected by Cell Counting Kit-8(CCK-8) assays and the cell morphology was determined by phalloidin staining. Osteogenetic genes expression levels were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. In addition, Autophagy-related genes expression levels were evaluated by immunostaining and western blotting in MC3T3-E1 in order to assess the induction of autophagy. RESULTS In this experiment, the 0.7 µm amplitude and 3 µm wavelength (W3) group increased the expression of osteogenic markers, whereas the 4.3 µm amplitude and 27 µm wavelength (W27) group showed inhibition. Both the cytoplasm and the nucleus of β-catenin, compared with those of the Flat, W3 increased, whereas W27 decreased. At the same time, the autophagy was consistent with the influence of the topography on osteogenic differentiation. Moreover, using CQ or RAPA significantly inhibited or promoted autophagy, as well as partially decreasing or increasing osteogenesis, respectively. Infecting siRNA-β-catenin decreased the expression of RUNX2 and OSX in MC3T3-E1 cells both treated with CQ and RAPA. CONCLUSIONS Wrinkled topographies activated the autophagy-mediated Wnt/β-catenin signaling pathway and affected the osteogenic differentiation of MC3T3-E1 cells. The introduction of aligned topographies on biomaterial scaffolds could provide physical cues with which modulate MC3T3-E1 responses for bone engineering constructs.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Qi Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education and Stomatological Hospital of Chongqing Medical University, Chongqing 401174, China
| | - Xiao Yan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Liping Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266003, China.
| | - Xiao Yuan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; School of Stomatology, Qingdao University, Qingdao 266003, China.
| |
Collapse
|
9
|
Peng G, Tsukamoto S, Ikutama R, Le Thanh Nguyen H, Umehara Y, Trujillo-Paez JV, Yue H, Takahashi M, Ogawa T, Kishi R, Tominaga M, Takamori K, Kitaura J, Kageyama S, Komatsu M, Okumura K, Ogawa H, Ikeda S, Niyonsaba F. Human-β-defensin-3 attenuates atopic dermatitis-like inflammation through autophagy activation and the aryl hydrocarbon receptor signaling pathway. J Clin Invest 2022; 132:156501. [PMID: 35834333 PMCID: PMC9435650 DOI: 10.1172/jci156501] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 07/12/2022] [Indexed: 01/18/2023] Open
Abstract
Human β-defensin-3 (hBD-3) exhibits antimicrobial and immunomodulatory activities; however, its contribution to autophagy regulation remains unclear, and the role of autophagy in the regulation of the epidermal barrier in atopic dermatitis (AD) is poorly understood. Here, keratinocyte autophagy was restrained in the skin lesions of patients with AD and murine models of AD. Interestingly, hBD-3 alleviated the IL-4– and IL-13–mediated impairment of the tight junction (TJ) barrier through keratinocyte autophagy activation, which involved aryl hydrocarbon receptor (AhR) signaling. While autophagy deficiency impaired the epidermal barrier and exacerbated inflammation, hBD-3 attenuated skin inflammation and enhanced the TJ barrier in AD. Importantly, hBD-3–mediated improvement of the TJ barrier was abolished in autophagy-deficient AD mice and in AhR-suppressed AD mice, suggesting a role for hBD-3–mediated autophagy in the regulation of the epidermal barrier and inflammation in AD. Thus, autophagy contributes to the pathogenesis of AD, and hBD-3 could be used for therapeutic purposes.
Collapse
Affiliation(s)
- Ge Peng
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saya Tsukamoto
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risa Ikutama
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hai Le Thanh Nguyen
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshie Umehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Juan V Trujillo-Paez
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hainan Yue
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miho Takahashi
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takasuke Ogawa
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryoma Kishi
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Mitsutoshi Tominaga
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Kenji Takamori
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Jiro Kitaura
- Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shun Kageyama
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigaku Ikeda
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Peng X, Ding C, Zhao Y, Hao M, Liu W, Yang M, Xiao F, Zheng Y. Poloxamer 407 and Hyaluronic Acid Thermosensitive Hydrogel-Encapsulated Ginsenoside Rg3 to Promote Skin Wound Healing. Front Bioeng Biotechnol 2022; 10:831007. [PMID: 35866029 PMCID: PMC9294355 DOI: 10.3389/fbioe.2022.831007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Ginsenoside Rg3 has shown beneficial effects in various skin diseases. The current interest in designing and developing hydrogels for biomedical applications continues to grow, inspiring the further development of drug-loaded hydrogels for tissue repair and localized drug delivery. The aim of the present study was to develop an effective and safe hydrogel (Rg3-Gel), using ginsenoside Rg3, and we evaluated the wound-healing potential and therapeutic mechanism of Rg3-Gel. The results indicated that the optimized Rg3-Gel underwent discontinuous phase transition at low and high temperatures. Rg3-Gel also exhibited good network structures, swelling water retention capacity, sustainable release performance, and excellent biocompatibility. Subsequently, the good antibacterial and antioxidant properties of Rg3-Gel were confirmed by in vitro tests. In full-thickness skin defect wounded models, Rg3-Gel significantly accelerated the wound contraction, promoted epithelial and tissue regeneration, and promoted collagen deposition and angiogenesis. In addition, Rg3-Gel increased the expression of autophagy proteins by inhibiting the MAPK and NF-KB pathways in vivo. It simultaneously regulated host immunity by increasing the abundance of beneficial bacteria and the diversity of the wound surface flora. From these preliminary evaluations, it is possible to conclude that Rg3-Gel has excellent application potential in wound-healing drug delivery systems.
Collapse
Affiliation(s)
- Xiaojuan Peng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Chuanbo Ding
- Jilin Agricultural Science and Technology University, Jilin, China
| | - Yingchun Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Mingqian Hao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Wencong Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- *Correspondence: Wencong Liu, ; Min Yang,
| | - Min Yang
- Jilin Agricultural Science and Technology University, Jilin, China
- *Correspondence: Wencong Liu, ; Min Yang,
| | - Fengyan Xiao
- Jilin Agricultural Science and Technology University, Jilin, China
| | - Yinan Zheng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| |
Collapse
|
11
|
LaRock DL, Johnson AF, Wilde S, Sands JS, Monteiro MP, LaRock CN. Group A Streptococcus induces GSDMA-dependent pyroptosis in keratinocytes. Nature 2022; 605:527-531. [PMID: 35545676 DOI: 10.1038/s41586-022-04717-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 04/01/2022] [Indexed: 12/21/2022]
Abstract
Gasdermins (GSDMs) are a family of pore-forming effectors that permeabilize the cell membrane during the cell death program pyroptosis1. GSDMs are activated by proteolytic removal of autoinhibitory carboxy-terminal domains, typically by caspase regulators1-9. However, no activator is known for one member of this family, GSDMA. Here we show that the major human pathogen group A Streptococcus (GAS) secretes a protease virulence factor, SpeB, that induces GSDMA-dependent pyroptosis. SpeB cleavage of GSDMA releases an active amino-terminal fragment that can insert into membranes to form lytic pores. GSDMA is primarily expressed in the skin10, and keratinocytes infected with SpeB-expressing GAS die of GSDMA-dependent pyroptosis. Mice have three homologues of human GSDMA, and triple-knockout mice are more susceptible to invasive infection by a pandemic hypervirulent M1T1 clone of GAS. These results indicate that GSDMA is critical in the immune defence against invasive skin infections by GAS. Furthermore, they show that GSDMs can act independently of host regulators as direct sensors of exogenous proteases. As SpeB is essential for tissue invasion and survival within skin cells, these results suggest that GSDMA can act akin to a guard protein that directly detects concerning virulence activities of microorganisms that present a severe infectious threat.
Collapse
Affiliation(s)
- Doris L LaRock
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Anders F Johnson
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Shyra Wilde
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Jenna S Sands
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Marcos P Monteiro
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA. .,Department of Medicine, Division of Infectious Diseases, Emory School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
12
|
Lavigne EG, Cavagnino A, Steinschneider R, Breton L, Baraibar MA, Jäger S. Oxidative damage prevention in human skin and sensory neurons by a salicylic acid derivative. Free Radic Biol Med 2022; 181:98-104. [PMID: 35114356 DOI: 10.1016/j.freeradbiomed.2022.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/18/2022] [Accepted: 01/30/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Increased protein carbonylation is a hallmark of oxidative stress, protein homeostasis dysregulation and aging in the nervous system and skin. Sensory neurons interact with skin cells and are involved in skin homeostasis. We have previously reported that the 5-octanoyl salicylic acid (C8-SA), a salicylic acid derivative, increased C. elegans lifespan and delayed the accumulation of carbonylated proteins, through the stimulation of autophagy. OBJECTIVES In this study we aimed to investigate if C8-SA protects human sensory neurons and human skin from extrinsic oxidative stressors as an approach to delay skin aging. METHODS In vitro reconstituted human epidermis innervated with hiPSc-derived human sensory neurons, as well as ex vivo human organotypic full skin models were used. The fully differentiated sensory neurons were pretreated with C8-SA before oxidative stress induction. Skin explants were maintained in culture and treated topically with C8-SA before the application of urban pollutants. Carbonylated proteins were detected using amino-oxy functionalized fluorophores and quantified. Chaperone mediated autophagy was monitored with LAMP2A immunofluorescence. Inflammation, ROS detoxification and autophagy were assessed by RT-PCR. RESULTS C8-SA prevented the accumulation of carbonylated proteins, both in human sensory neurons and skin explants. C8-SA stimulated chaperone-mediated autophagy and modulated NRF2 antioxidant response genes, as well as catalase enzymatic activity. CONCLUSIONS C8-SA acts at two levels to protect skin against oxidative stress: 1) it prevents protein oxidation by stimulating endogenous antioxidant defense and 2) it increases the clearance of oxidized proteins by stimulating chaperone-mediated autophagy. These results suggest that C8-SA maintains skin health in urban polluted environments.
Collapse
Affiliation(s)
| | | | | | - Lionel Breton
- L'OREAL Research &Innovation, Aulnay-sous-bois, France
| | | | - Sibylle Jäger
- L'OREAL Research &Innovation, Aulnay-sous-bois, France.
| |
Collapse
|
13
|
Fabrication of chitosan/PVP/dihydroquercetin nanocomposite film for in vitro and in vivo evaluation of wound healing. Int J Biol Macromol 2022; 206:591-604. [PMID: 35217084 DOI: 10.1016/j.ijbiomac.2022.02.110] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
The high cost of wound healing treatment, the slow recovery of wounds, and the uncertainty of being affected by the body's physiological activities constitute a serious burden on public health. In this work, we report the preparation and characterization of chitosan (CS), PVP, and dihydroquercetin (DHQ) nanofiber film used as wound excipients, as well as in vivo and in vitro evaluations, and verify that the film is effective in wounds. The results show that the prepared film has good morphology, thermal stability and hydrophilicity. In vitro studies have shown that it has antibacterial activity against S.aureus and E.coli, and the DPPH free radical scavenging rate proves that the fiber film has antioxidant activity. MTT cytotoxicity test proved that the film is non-toxic to Hacat cells. Animal experiments have proved that wounds treated with CS-PVP-DHQ nanofiber film heal faster. This article also studied the composite nanofiber film by inducing autophagy pathway and increasing the expression of pan-keratin, vascular endothelial growth factor VEGF and CD31 to promote wound healing. Therefore, the nanofiber film herein show great potential in wound healing applications.
Collapse
|
14
|
Ripszky Totan A, Greabu M, Stanescu-Spinu II, Imre M, Spinu TC, Miricescu D, Ilinca R, Coculescu EC, Badoiu SC, Coculescu BI, Albu C. The Yin and Yang dualistic features of autophagy in thermal burn wound healing. Int J Immunopathol Pharmacol 2022; 36:3946320221125090. [PMID: 36121435 PMCID: PMC9490459 DOI: 10.1177/03946320221125090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Burn healing should be regarded as a dynamic process consisting of two main, interrelated phases: (a) the inflammatory phase when neutrophils and monocytes infiltrate the injury site, through localized vasodilation and fluid extravasation, and (b) the proliferative-remodeling phase, which represents a key event in wound healing. In the skin, both canonical autophagy (induced by starvation, oxidative stress, and environmental aggressions) and non-canonical or selective autophagy have evolved to play a discrete, but, essential, “housekeeping” role, for homeostasis, immune tolerance, and survival. Experimental data supporting the pro-survival roles of autophagy, highlighting its Yang, luminous and positive feature of this complex but insufficient explored molecular pathway, have been reported. Autophagic cell death describes an “excessive” degradation of important cellular components that are necessary for normal cell function. This deadly molecular mechanism brings to light the darker, concealed, Yin feature of autophagy. Autophagy seems to perform dual, conflicting roles in the angiogenesis context, revealing once again, its Yin–Yang features. Autophagy with its Yin–Yang features remains the shadow player, able to decide quietly whether the cell survives or dies.
Collapse
Affiliation(s)
- Alexandra Ripszky Totan
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Maria Greabu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Iulia-Ioana Stanescu-Spinu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Marina Imre
- Department of Complete Denture, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Tudor-Claudiu Spinu
- Department of Fixed Prosthodontics and Occlusology, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Daniela Miricescu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Radu Ilinca
- Department of Biophysics, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Elena Claudia Coculescu
- Department of Oral Pathology, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Silviu Constantin Badoiu
- Department of Anatomy and Embryology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan-Ioan Coculescu
- Cantacuzino National Medico-Military Institute for Research and Development, Bucharest, Romania
| | - Crenguta Albu
- Department of Genetics, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Bucharest, Romania
| |
Collapse
|
15
|
Liu H, Huang Y, Yang Y, Han Y, Jia L, Li W. Compressive force-induced LincRNA-p21 inhibits mineralization of cementoblasts by impeding autophagy. FASEB J 2021; 36:e22120. [PMID: 34958157 DOI: 10.1096/fj.202101589r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/30/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
The mineralization capability of cementoblasts is the foundation for repairing orthodontic treatment-induced root resorption. It is essential to investigate the regulatory mechanism of mineralization in cementoblasts under mechanical compression to improve orthodontic therapy. Autophagy has a protective role in maintaining cell homeostasis under environmental stress and was reported to be involved in the mineralization process. Long noncoding RNAs are important regulators of biological processes, but their functions in compressed cementoblasts during orthodontic tooth movement remain unclear. In this study, we showed that compressive force downregulated the expression of mineralization-related markers. LincRNA-p21 was strongly enhanced by compressive force. Overexpression of lincRNA-p21 downregulated the expression of mineralization-related markers, while knockdown of lincRNA-p21 reversed the compressive force-induced decrease in mineralization. Furthermore, we found that autophagy was impeded in compressed cementoblasts. Then, overexpression of lincRNA-p21 decreased autophagic activity, while knockdown of lincRNA-p21 reversed the autophagic process decreased by mechanical compression. However, the autophagy inhibitor 3-methyladenine abolished the lincRNA-p21 knockdown-promoted mineralization, and the autophagy activator rapamycin rescued the mineralization inhibited by lincRNA-p21 overexpression. Mechanistically, the direct binding between lincRNA-p21 and FoxO3 blocked the expression of autophagy-related genes. In a mouse orthodontic tooth movement model, knockdown of lincRNA-p21 rescued the impeded autophagic process in cementoblasts, enhanced cementogenesis, and alleviated orthodontic force-induced root resorption. Overall, compressive force-induced lincRNA-p21 inhibits the mineralization capability of cementoblasts by impeding the autophagic process.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yiping Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuhui Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yineng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lingfei Jia
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
16
|
Lefèvre-Utile A, Braun C, Haftek M, Aubin F. Five Functional Aspects of the Epidermal Barrier. Int J Mol Sci 2021; 22:11676. [PMID: 34769105 PMCID: PMC8583944 DOI: 10.3390/ijms222111676] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023] Open
Abstract
The epidermis is a living, multilayered barrier with five functional levels, including a physical, a chemical, a microbial, a neuronal, and an immune level. Altogether, this complex organ contributes to protect the host from external aggression and to preserve its integrity. In this review, we focused on the different functional aspects.
Collapse
Affiliation(s)
- Alain Lefèvre-Utile
- Sce de Pédiatrie Générale et Urgence pédiatrique, Hôpital Jean Verdier, Assistance Publique Hôpitaux de Paris, 93140 Bondy, France;
- Unité 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Inserm, 75010, Paris, France
| | - Camille Braun
- Centre international de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, 69007 Lyon, France;
- Sce de Pneumologie Pédiatrique et Allergie, Hôpital Femme Mère Enfant, 69500 Bron, France
| | - Marek Haftek
- CNRS UMR5305, Laboratory of Tissue Biology and Therapeutic Engineering, LBTI, Lyon1 University, 69100 Lyon, France;
| | - François Aubin
- Inserm U1098, Université de Franche Comté, 25000 Besançon, France
- Sce de Dermatologie, Centre Hospitalier Universitaire, 25000 Besançon, France
| |
Collapse
|
17
|
Liang D, Lin WJ, Ren M, Qiu J, Yang C, Wang X, Li N, Zeng T, Sun K, You L, Yan L, Wang W. m 6A reader YTHDC1 modulates autophagy by targeting SQSTM1 in diabetic skin. Autophagy 2021; 18:1318-1337. [PMID: 34657574 PMCID: PMC9225222 DOI: 10.1080/15548627.2021.1974175] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dysregulation of macroautophagy/autophagy contributes to the delay of wound healing in diabetic skin. N6-methyladenosine (m6A) RNA modification is known to play a critical role in regulating autophagy. In this study, it was found that SQSTM1/p62 (sequestosome 1), an autophagy receptor, was significantly downregulated in two human keratinocyte cells lines with short-term high-glucose treatment, as well as in the epidermis of diabetic patients and a db/db mouse model with long-term hyperglycemia. Knockdown of SQSTM1 led to the impairment of autophagic flux, which was consistent with the results of high-glucose treatment in keratinocytes. Moreover, the m6A reader protein YTHDC1 (YTH domain containing 1), which interacted with SQSTM1 mRNA, was downregulated in keratinocytes under both the acute and chronic effects of hyperglycemia. Knockdown of YTHDC1 affected biological functions of keratinocytes, which included increased apoptosis rates and impaired wound-healing capacity. In addition, knockdown of endogenous YTHDC1 resulted in a blockade of autophagic flux in keratinocytes, while overexpression of YTHDC1 rescued the blockade of autophagic flux induced by high glucose. In vivo, knockdown of endogenous Ythdc1 or Sqstm1 inhibited autophagy in the epidermis and delayed wound healing. Interestingly, we found that a decrease of YTHDC1 drove SQSTM1 mRNA degradation in the nucleus. Furthermore, the results revealed that YTHDC1 interacted and cooperated with ELAVL1/HuR (ELAV like RNA binding protein 1) in modulating the expression of SQSTM1. Collectively, this study uncovered a previously unrecognized function for YTHDC1 in modulating autophagy via regulating the stability of SQSTM1 nuclear mRNA in diabetic keratinocytes. Abbreviations: ACTB: actin beta; AGEs: glycation end products; AL: autolysosome; AP: autophagosome; ATG: autophagy related; AKT: AKT serine/threonine kinase; ANOVA: analysis of variance; BECN1: beclin 1; Co-IP: co-immunoprecipitation; DEGs: differentially expressed genes; DM: diabetes mellitus; ELAVL1: ELAV like RNA binding protein 1; FTO: FTO alpha-ketoglutarate dependent dioxygenase; G: glucose; HaCaT: human keratinocyte; GO: Gene Ontology; GSEA: Gene Set Enrichment Analysis; HE: hematoxylin-eosin; IHC: immunohistochemical; IRS: immunoreactive score; KEAP1: kelch like ECH associated protein 1; KEGG: Kyoto Encyclopedia of Genes and Genomes; m6A: N6-methyladenosine; M: mannitol; MANOVA: multivariate analysis of variance; MAP1LC3: microtubule associated protein 1 light chain 3; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MeRIP: methylated RNA immunoprecipitation; METTL3: methyltransferase 3, N6-adenosine-methytransferase complex catalytic subunit; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NBR1: NBR1 autophagy cargo receptor; NFE2L2: nuclear factor, erythroid 2 like 2; NG: normal glucose; NHEK: normal human epithelial keratinocyte; OE: overexpressing; p-: phospho-; PI: propidium iodide; PPIN: Protein-Protein Interaction Network; RBPs: RNA binding proteins; RIP: RNA immunoprecipitation; RNA-seq: RNA-sequence; RNU6–1: RNA, U6 small nuclear 1; ROS: reactive oxygen species; siRNAs: small interfering RNAs; SQSTM1: sequestosome 1; SRSF: serine and arginine rich splicing factor; T2DM: type 2 diabetes mellitus; TEM: transmission electron microscopy; TUBB: tubulin beta class I; WT: wild-type; YTHDC1: YTH domain containing 1.
Collapse
Affiliation(s)
- Diefei Liang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center of Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meng Ren
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junxiong Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuan Yang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Na Li
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tingting Zeng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kan Sun
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lili You
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Hill D, Cosgarea I, Reynolds N, Lovat P, Armstrong J. Research Techniques Made Simple: Analysis of Autophagy in the Skin. J Invest Dermatol 2021; 141:5-9.e1. [PMID: 33342508 DOI: 10.1016/j.jid.2020.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/21/2020] [Accepted: 10/11/2020] [Indexed: 02/05/2023]
Abstract
Autophagy is required for normal skin homeostasis and its disordered regulation is implicated in a range of cutaneous diseases. Several well-characterized biomarkers of autophagy are used experimentally to quantify autophagic activity or clinically to correlate autophagy with disease progression. This article discusses the advantages and limitations of different approaches for measuring autophagy as well as the techniques for modulating autophagy. These include analysis of endogenous LC3, a central autophagy regulatory protein, and measurement of LC3 flux using a dual-fluorescent reporter, which provides a quantitative readout of autophagy in cell culture systems in vitro and animal models in vivo. Degradation of SQSTM1/p62 during autophagy is proposed as an alternative biomarker allowing the analysis of autophagy both experimentally and clinically. However, the complex regulation of individual autophagy proteins and their involvement in multiple pathways means that several proteins must be analyzed together, preferably over a time course to accurately interpret changes in autophagic activity. Genetic modification of autophagy proteins can be used to better understand basic autophagic mechanisms contributing to health and disease, whereas small molecule inhibitors of autophagy regulatory proteins, lysosomal inhibitors, or activators of cytotoxic autophagy have been explored as potential treatments for skin disorders where autophagy is defective.
Collapse
Affiliation(s)
- David Hill
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom
| | - Ioana Cosgarea
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle Dermatology, Newcastle Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom; Newcastle Oncology, Newcastle Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Nick Reynolds
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle Dermatology, Newcastle Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Penny Lovat
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; AMLo Biosciences Ltd, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jane Armstrong
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom.
| |
Collapse
|
19
|
Choi MS, Chae YJ, Choi JW, Chang JE. Potential Therapeutic Approaches through Modulating the Autophagy Process for Skin Barrier Dysfunction. Int J Mol Sci 2021; 22:7869. [PMID: 34360634 PMCID: PMC8345957 DOI: 10.3390/ijms22157869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Autophagy is an attractive process to researchers who are seeking novel potential treatments for various diseases. Autophagy plays a critical role in degrading damaged cellular organelles, supporting normal cell development, and maintaining cellular homeostasis. Because of the various effects of autophagy, recent human genome research has focused on evaluating the relationship between autophagy and a wide variety of diseases, such as autoimmune diseases, cancers, and inflammatory diseases. The skin is the largest organ in the body and provides the first line of defense against environmental hazards, including UV damage, chemical toxins, injuries, oxidative stress, and microorganisms. Autophagy takes part in endogenous defense mechanisms by controlling skin homeostasis. In this manner, regulating autophagy might contribute to the treatment of skin barrier dysfunctions. Various studies are ongoing to elucidate the association between autophagy and skin-related diseases in order to find potential therapeutic approaches. However, little evidence has been gathered about the relationship between autophagy and the skin. In this review, we highlight the previous findings of autophagy and skin barrier disorders and suggest potential therapeutic strategies. The recent research regarding autophagy in acne and skin aging is also discussed.
Collapse
Affiliation(s)
- Min Sik Choi
- Lab of Pharmacology, College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea;
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju-gun 55338, Korea;
| | - Ji Woong Choi
- College of Pharmacy, Gachon University, Incheon 21936, Korea;
| | - Ji-Eun Chang
- Lab of Pharmaceutics, College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea
| |
Collapse
|
20
|
Margaritte-Jeannin P, Budu-Aggrey A, Ege M, Madore AM, Linhard C, Mohamdi H, von Mutius E, Granell R, Demenais F, Laprise C, Bouzigon E, Dizier MH. Identification of OCA2 as a novel locus for the co-morbidity of asthma-plus-eczema. Clin Exp Allergy 2021; 52:70-81. [PMID: 34155719 DOI: 10.1111/cea.13972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Numerous genes have been associated with the three most common allergic diseases (asthma, allergic rhinitis or eczema) but these genes explain only a part of the heritability. In the vast majority of genetic studies, complex phenotypes such as co-morbidity of two of these diseases, have not been considered. This may partly explain missing heritability. OBJECTIVE To identify genetic variants specifically associated with the co-morbidity of asthma-plus-eczema. METHODS We first conducted a meta-analysis of four GWAS (Genome-Wide Association Study) of the combined asthma-plus-eczema phenotype (total of 8807 European-ancestry subjects of whom 1208 subjects had both asthma and eczema). To assess whether the association with SNP(s) was specific to the co-morbidity, we also conducted a meta-analysis of homogeneity test of association according to disease status ("asthma-plus-eczema" vs. the presence of only one disease "asthma only or eczema only"). We then used a joint test by combining the two test statistics from the co-morbidity-SNP association and the phenotypic heterogeneity of SNP effect meta-analyses. RESULTS Seven SNPs were detected for specific association to the asthma-plus-eczema co-morbidity, two with significant and five with suggestive evidence using the joint test after correction for multiple testing. The two significant SNPs are located in the OCA2 gene (Oculocutaneous Albinism II), a new locus never detected for significant evidence of association with any allergic disease. This gene is a promising candidate gene, because of its link to skin and lung diseases, and to epithelial barrier and immune mechanisms. CONCLUSION Our study underlines the importance of studying sub-phenotypes as co-morbidities to detect new susceptibility genes.
Collapse
Affiliation(s)
| | - Ashley Budu-Aggrey
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Markus Ege
- Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Dr von Hauner Children's Hospital, Ludwig Maximilian University, Munich, Germany
| | - Anne-Marie Madore
- Département des Sciences Fondamentales, Centre Intersectoriel en Santé Durable (CISD), Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | | | | | - Erika von Mutius
- Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Dr von Hauner Children's Hospital, Ludwig Maximilian University, Munich, Germany
| | - Raquel Granell
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Catherine Laprise
- Département des Sciences Fondamentales, Centre Intersectoriel en Santé Durable (CISD), Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | | | | |
Collapse
|
21
|
Jeong D, Qomaladewi NP, Lee J, Park SH, Cho JY. The Role of Autophagy in Skin Fibroblasts, Keratinocytes, Melanocytes, and Epidermal Stem Cells. J Invest Dermatol 2021; 140:1691-1697. [PMID: 32800183 DOI: 10.1016/j.jid.2019.11.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 10/30/2019] [Accepted: 11/09/2019] [Indexed: 12/20/2022]
Abstract
Human skin acts as a barrier to protect our bodies from UV rays and external pathogens and to prevent water loss. Phenotypes of aging, or natural aging due to chronic damage, include wrinkles and the reduction of skin thickness that occur because of a loss of skin cell function. The dysregulation of autophagy, a lysosome-related degradation pathway, can lead to cell senescence, cancer, and various human diseases due to abnormal cellular homeostasis. Here, we discuss the roles and molecular mechanisms of autophagy involved in the anti-aging effects of autophagy and the relationship between autophagy and aging in skin cells.
Collapse
Affiliation(s)
- Deok Jeong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea
| | | | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon, Korea
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon, Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon, Korea.
| |
Collapse
|
22
|
Simpson CL, Tokito MK, Uppala R, Sarkar MK, Gudjonsson JE, Holzbaur ELF. NIX initiates mitochondrial fragmentation via DRP1 to drive epidermal differentiation. Cell Rep 2021; 34:108689. [PMID: 33535046 PMCID: PMC7888979 DOI: 10.1016/j.celrep.2021.108689] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/23/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
The epidermis regenerates continually to maintain a protective barrier at the body’s surface composed of differentiating keratinocytes. Maturation of this stratified tissue requires that keratinocytes undergo wholesale organelle degradation upon reaching the outermost tissue layers to form compacted, anucleate cells. Through live imaging of organotypic cultures of human epidermis, we find that regulated breakdown of mitochondria is critical for epidermal development. Keratinocytes in the upper layers initiate mitochondrial fragmentation, depolarization, and acidification upon upregulating the mitochondrion-tethered autophagy receptor NIX. Depleting NIX compromises epidermal maturation and impairs mitochondrial elimination, whereas ectopic NIX expression accelerates keratinocyte differentiation and induces premature mitochondrial fragmentation via the guanosine triphosphatase (GTPase) DRP1. We further demonstrate that inhibiting DRP1 blocks NIX-mediated mitochondrial breakdown and disrupts epidermal development. Our findings establish mitochondrial degradation as a key step in terminal keratinocyte differentiation and define a pathway operating via the mitophagy receptor NIX in concert with DRP1 to drive epidermal morphogenesis. Using live microscopy of human organotypic epidermis, Simpson et al. demonstrate how keratinocytes degrade their mitochondria in the upper tissue layers during their final stage of differentiation. By upregulating expression of the mitophagy receptor NIX, keratinocytes initiate DRP1- dependent mitochondrial fragmentation, a process critical for epidermal tissue maturation.
Collapse
Affiliation(s)
- Cory L Simpson
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mariko K Tokito
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ranjitha Uppala
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Breda LCD, Menezes IG, Paulo LNM, de Almeida SR. Immune Sensing and Potential Immunotherapeutic Approaches to Control Chromoblastomycosis. J Fungi (Basel) 2020; 7:jof7010003. [PMID: 33375204 PMCID: PMC7822212 DOI: 10.3390/jof7010003] [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] [Received: 11/19/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022] Open
Abstract
Chromoblastomycosis (CBM) is a neglected, chronic, and progressive subcutaneous mycosis caused by different species of fungi from the Herpotrichiellaceae family. CBM disease is usually associated with agricultural activities, and its infection is characterized by verrucous, erythematous papules, and atrophic lesions on the upper and lower limbs, leading to social stigma and impacts on patients' welfare. The economic aspect of disease treatment is another relevant issue. There is no specific treatment for CBM, and different anti-fungal drug associations are used to treat the patients. However, the long period of the disease and the high cost of the treatment lead to treatment interruption and, consequently, relapse of the disease. In previous years, great progress had been made in the comprehension of the CBM pathophysiology. In this review, we discuss the differences in the cell wall composition of conidia, hyphae, and muriform cells, with a particular focus on the activation of the host immune response. We also highlight the importance of studies about the host skin immunology in CBM. Finally, we explore different immunotherapeutic studies, highlighting the importance of these approaches for future treatment strategies for CBM.
Collapse
|
24
|
Lee Y, Shin K, Shin KO, Yoon S, Jung J, Hwang E, Chung HJ, Hossini AM, Zouboulis CC, Baek MJ, Baek JH, Chi YM, Lee S, Jeong S. Topical application of autophagy-activating peptide improved skin barrier function and reduced acne symptoms in acne-prone skin. J Cosmet Dermatol 2020; 20:1009-1016. [PMID: 32697858 DOI: 10.1111/jocd.13636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent studies about the important roles of autophagy signaling in sebaceous lipogenesis and epidermal differentiation suggest potential benefits of autophagy activation in acne. AIMS To investigate the effects of an autophagy activator on acne-prone skin. METHODS Autophagy signaling in human immortalized SZ95 sebocytes, normal human epidermal keratinocytes, and 3D reconstituted skin was examined. Effects of an autophagy-activating peptide on sebaceous lipogenesis were measured by fluorescence microscopic analysis. The clinical efficacy in acne-prone skin was evaluated through an eight-week, double-blind, randomized, vehicle-controlled study. Changes in skin surface lipid compositions were further analyzed. RESULTS In cultured sebocytes and keratinocytes, the investigated autophagy-activating peptide increased LC3-II expression, indicating a stimulation of autophagy signaling. Testosterone and linoleic acid treatment induced lipogenesis in cultured sebocytes and is further inhibited by the autophagy activator peptide treatment. Increased expression of differentiation marker proteins in cultured keratinocytes was also observed by autophagy-activating peptide. In clinical study, reduction of closed comedones and the amount of skin surface lipids as well as of trans-epidermal water loss (TEWL) were observed in acne-prone skin after autophagy-activating peptide application. In addition, reduction of squalene and increase in cholesterol were observed after an 8-week application. CONCLUSIONS Topical application of an autophagy activator downregulated sebaceous lipogenesis and improved the skin barrier function. Considering the important roles of sebum and skin barrier function in acne pathogenesis, autophagy activation might represent a new therapeutic option in early forms of acne.
Collapse
Affiliation(s)
- Yoonjin Lee
- College of Life Science and Biotechnology, Korea University, Seoul, South Korea
| | - Kayoung Shin
- Research Division, Incospharm Corp. Daejeon, South Korea
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Seokjeong Yoon
- Research Division, Incospharm Corp. Daejeon, South Korea
| | - Juyeon Jung
- Research Division, Incospharm Corp. Daejeon, South Korea
| | - Eojin Hwang
- Research Division, Incospharm Corp. Daejeon, South Korea
| | - Hwa-Jee Chung
- Research Division, Incospharm Corp. Daejeon, South Korea
| | - Amir M Hossini
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, Dessau, Germany
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, Dessau, Germany
| | - Min Jeong Baek
- Dermapro Skin Research Center, DERMAPRO Ltd., Seoul, South Korea
| | - Ji Hwoon Baek
- Dermapro Skin Research Center, DERMAPRO Ltd., Seoul, South Korea
| | - Young Min Chi
- College of Life Science and Biotechnology, Korea University, Seoul, South Korea
| | - Sangeun Lee
- Department of Dermatology, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Korea
| | - Sekyoo Jeong
- Research Division, Incospharm Corp. Daejeon, South Korea
| |
Collapse
|
25
|
Sil P, Suwanpradid J, Muse G, Gruzdev A, Liu L, Corcoran DL, Willson CJ, Janardhan K, Grimm S, Myers P, Degraff LM, MacLeod AS, Martinez J. Noncanonical autophagy in dermal dendritic cells mediates immunosuppressive effects of UV exposure. J Allergy Clin Immunol 2019; 145:1389-1405. [PMID: 31837371 DOI: 10.1016/j.jaci.2019.11.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/10/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Control of the inflammatory response is critical to maintaining homeostasis, and failure to do so contributes to the burden of chronic inflammation associated with several disease states. The mechanisms that underlie immunosuppression, however, remain largely unknown. Although defects in autophagy machinery have been associated with inflammatory pathologic conditions, we now appreciate that autophagic components participate in noncanonical pathways distinct from classical autophagy. We have previously demonstrated that LC3-associated phagocytosis (LAP), a noncanonical autophagic process dependent on Rubicon (rubicon autophagy regulator [RUBCN]), contributes to immunosuppression. OBJECTIVE We used Rubcn-/- mice to examine the role of the LAP pathway in mediating the UV-induced immunotolerant program in a model of contact hypersensitivity (CHS). METHODS Flow cytometry and transcriptional analysis were used to measure immune cell infiltration and activation in the skin of Rubcn+/+ and Rubcn-/- mice during the CHS response. RESULTS Here, we demonstrate that LAP is required for UV-induced immunosuppression and that UV exposure induces a broadly anti-inflammatory transcriptional program dependent on Rubicon. Rubcn-/- mice are resistant to UV-induced immunosuppression and instead display exaggerated inflammation in a model of CHS. Specifically, RUBCN deficiency in CD301b+ dermal dendritic cells results in their increased antigen presentation capacity and subsequent hyperactivation of the CD8+ T-cell response. CONCLUSIONS LAP functions to limit the immune response and is critical in maintaining the balance between homeostasis and inflammation.
Collapse
Affiliation(s)
- Payel Sil
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | | | - Ginger Muse
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Liwen Liu
- Molecular Genomics Core Laboratory, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - David L Corcoran
- Duke Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC
| | | | | | - Sara Grimm
- Division of Intramural Research, Research Triangle Park, NC
| | - Page Myers
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Laura Miller Degraff
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC; Department of Immunology, Duke University, Durham, NC; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
| |
Collapse
|
26
|
Autophagy: Multiple Mechanisms to Protect Skin from Ultraviolet Radiation-Driven Photoaging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8135985. [PMID: 31915514 PMCID: PMC6930764 DOI: 10.1155/2019/8135985] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/26/2019] [Indexed: 01/07/2023]
Abstract
Autophagy is an essential cellular process that maintains balanced cell life. Restriction in autophagy may induce degenerative changes in humans. Natural or pathological aging of susceptible tissues has been linked with reduced autophagic activity. Skin photoaging is an example of such pathological condition caused by ambient solar UV radiation exposure. The UV-induced production of reaction oxygen species (ROS) has been linked to the promotion and progression of the photoaging process in exposed tissues. Accordingly, it has been suggested that autophagy is capable of delaying the skin photoaging process caused by solar ultraviolet (UV), although the underlying mechanism is still under debate. This review highlights several plausible mechanisms by which UV-induced ROS activates the cellular signaling pathways and modulates the autophagy. More specifically, the UV-mediated regulation of autophagy and age-related transcription factors is discussed to pinpoint the contribution of autophagy to antiphotoaging effects in the skin. The outcome of this review will provide insights into design intervention strategies for delaying the phenomenon of sunlight-induced photodamage, photoaging, and other aging-related chronic diseases based on factors that activate the autophagy process in the skin.
Collapse
|
27
|
Rabiee Motmaen S, Tavakol S, Joghataei MT, Barati M. Acidic pH derived from cancer cells as a double-edged knife modulates wound healing through DNA repair genes and autophagy. Int Wound J 2019; 17:137-148. [PMID: 31714008 DOI: 10.1111/iwj.13248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 12/16/2022] Open
Abstract
Wound healing is a sequester program that involves diverse cell signalling cascades. Notwithstanding, complete signal transduction pathways underpinning acidic milieu derived from cancer cells is not clear, yet. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, fluorescein diacetate/propidium iodide staining, and cell cycle flow cytometry revealed that acidic media decreased cell viability and cell number along with enhanced dead cells and S-phase arrest in normal fibroblasts. Notably, the trends of intracellular reactive oxygen species production and lactate dehydrogenase release significantly increased with time. It seems the downregulation of Klf4 is in part due to acidosis-induced DNA damage. It promoted cells towards S-phase arrest and diminished cell proliferation. Klf4 downregulation had a direct correlation with the P53 level while acidic microenvironment promotes cells towards cell death mechanisms including apoptosis and autophagy. Noteworthily, the unchanged levels of Rb and Mlh1 indicated in those genes had no dominant role in the repairing of DNA damage in fibroblasts treated with the acidic microenvironment. Therefore, cells owing to not entering to mitosis and accumulation of DNA damage were undergone cell death to preserve cell homeostasis. Since acidic media decreased the level of tumour suppressor and DNA repair genes and altered the normal survival pathways in fibroblasts, caution should be exercised to not lead to cancer rather than wound healing.
Collapse
Affiliation(s)
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad T Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Barati
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Li L, Yang S, Xu L, Li Y, Fu Y, Zhang H, Song J. Nanotopography on titanium promotes osteogenesis via autophagy-mediated signaling between YAP and β-catenin. Acta Biomater 2019; 96:674-685. [PMID: 31284094 DOI: 10.1016/j.actbio.2019.07.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/17/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
Nanostructured titanium implants are recognized for inducing osteogenesis, but the cell signal transductions related to topography are not fully understood. Implant topography is associated with the functionality of osteogenic transcription factors directed by β-catenin in the nucleus, and autophagic flux in the cytoplasm; YAP (Yes-associated protein) is implicated in the destruction of β-catenin in the cytoplasm and is susceptible to autophagic flux. This study investigated whether surface topography of the titanium implant modulates autophagy-lysosome degradation of cytoplasmic YAP. Titanium surfaces were modified with smooth, micro, or nanotopographies. Compared with the smooth and micro surfaces, nanotopography was associated with higher β-catenin nuclear translocation, osteogenic differentiation, and autophagy, and less cytoplasmic YAP. Blockade of the autophagy-lysosome pathway resulted in YAP retention in MC3T3-E1 cells. Cytoplasmic YAP restricted β-catenin nuclear translocation. In the nano surface group, β-catenin accumulation in the nucleus and expression of osteogenesis genes was improved. However, in the absence of cell-cell (confluent) contact, manipulation of YAP and β-catenin localization associated with topography-induced autophagy was lost. In summary, the osteogenesis observed in response to titanium implants with nanotopography involves a signaling link between YAP and β-catenin. STATEMENT OF SIGNIFICANCE: Titanium with rough topographical surfaces is extensively applied in orthopedic and dental clinics. However, the cellular response to topographies that promotes osteogenesis and underlying mechanisms are not fully understood. In this study, we modified titanium surfaces to produce smooth, micro, or nano topographies. Experiments indicated that the nanotopography induced a stronger autophagic response, leading to degraded cytoplasmic YAP. With the lower levels of YAP, β-catenin transported and accumulated in the nucleus to activate TCF/LEF transcription factors, resulting in stronger osteogenesis. Additionally, cell-cell contact was essential in the autophagy-mediated signaling link between YAP and β-catenin. Consequently, our investigation revealed a novel signal transduction in nanotopography-regulated osteogenesis, and supports the modification of biomaterial surfaces to maximize osseointegration.
Collapse
Affiliation(s)
- Lingjie Li
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Sheng Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Ling Xu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Yuzhou Li
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Yiru Fu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - He Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| |
Collapse
|
29
|
Eckhart L, Tschachler E, Gruber F. Autophagic Control of Skin Aging. Front Cell Dev Biol 2019; 7:143. [PMID: 31417903 PMCID: PMC6682604 DOI: 10.3389/fcell.2019.00143] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
The skin forms the barrier to the environment. Maintenance of this barrier during aging requires orchestrated responses to variable types of stress, the continuous renewal of the epithelial compartment, and the homeostasis of long-lived cell types. Recent experimental evidence suggests that autophagy is critically involved in skin homeostasis and skin aging is associated with and partially caused by defects of autophagy. In the outer skin epithelium, autophagy is constitutively active during cornification of keratinocytes and increases the resistance to environmental stress. Experimental suppression of autophagy in the absence of stress is tolerated by the rapidly renewing epidermal epithelium, whereas long-lived skin cells such as melanocytes, Merkel cells and secretory cells of sweat glands depend on autophagy for cellular homeostasis and normal execution of their functions during aging. Yet other important roles of autophagy have been identified in the dermis where senescence of mesenchymal cells and alterations of the extracellular matrix (ECM) are hallmarks of aging. Here, we review the evidence for cell type-specific roles of autophagy in the skin and their differential contributions to aging.
Collapse
Affiliation(s)
- Leopold Eckhart
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Florian Gruber
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
| |
Collapse
|
30
|
Gramlich R, Aliahmadi E, Peiser M. In Vitro Induction of T Helper 17 Cells by Synergistic Activation of Human Monocyte-Derived Langerhans Cell-Like Cells with Bacterial Agonists. Int J Mol Sci 2019; 20:ijms20061367. [PMID: 30893757 PMCID: PMC6471444 DOI: 10.3390/ijms20061367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/02/2023] Open
Abstract
In the case of epidermal barrier disruption, pathogens encounter skin-resident Langerhans cells (LCs) and are recognized by pathogen recognition receptors such as Toll-like receptors (TLRs). As the majority of microorganisms exhibit more than one TLR ligand, the mechanisms of subsequent T cell differentiation are complex and far from clear. In this study, we investigated combinatory effects on Th cell polarization by bacterial cell wall compounds peptidoglycan (PGN) and lipopolysaccharide (LPS) and by bacterial nucleic acid (DNA). Expression of maturation markers CD40, CD80, HLA-DR and CCR7 and the release of IL-1β, IL-6 and IL-23 was strongly enhanced by simultaneous exposure to PGN, LPS and DNA in LCs. As all these factors were potential Th17 driving cytokines, we investigated the potency of combinatory TLR stimuli to induce Th17 cells via LC activation. High amounts of IL-17A and IL-22, key cytokines of Th17 cells, were detected. By intracellular costaining of IL-17+T cells, IL-22− (Th17) and IL-22+ (immature Th17) cells were identified. Interestingly, one population of LPS stimulated cells skewed into IL-9+Th cells, and LPS synergized with PGN while inducing high IL-22. In conclusion, our data indicates that when mediated by a fine-tuned signal integration via LCs, bacterial TLR agonists synergize and induce Th17 differentiation.
Collapse
Affiliation(s)
- Robert Gramlich
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Center for Anesthesiology and Intensive Care Medicine, Department of intensive Care, Medical Center Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246 Hamburg, Germany.
| | - Ehsan Aliahmadi
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Surgery and Vascular Surgery, Franziskus Krankenhaus Berlin, University Hospital of Charité Universitätsmedizin Berlin, Budapester Straße 15-19, 10787 Berlin, Germany.
| | - Matthias Peiser
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany.
| |
Collapse
|
31
|
Autophagy Activation by Crepidiastrum Denticulatum Extract Attenuates Environmental Pollutant-Induced Damage in Dermal Fibroblasts. Int J Mol Sci 2019; 20:ijms20030517. [PMID: 30691106 PMCID: PMC6386979 DOI: 10.3390/ijms20030517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 12/12/2022] Open
Abstract
Pollution-induced skin damage results in oxidative stress; cellular toxicity; inflammation; and, ultimately, premature skin aging. Previous studies suggest that the activation of autophagy can protect oxidation-induced cellular damage and aging-like changes in skin. In order to develop new anti-pollution ingredients, this study screened various kinds of natural extracts to measure their autophagy activation efficacy in cultured dermal fibroblast. The stimulation of autophagy flux by the selected extracts was further confirmed both by the expression of proteins associated with the autophagy signals and by electron microscope. Crepidiastrum denticulatum (CD) extract treated cells showed the highest autophagic vacuole formation in the non-cytotoxic range. The phosphorylation of adenosine monophosphate kinase (AMPK), but not the inhibition of mammalian target of rapamycin (mTOR), was observed by CD-extract treatment. Its anti-pollution effects were further evaluated with model compounds, benzo[a]pyrene (BaP) and cadmium chloride (CdCl2), and a CD extract treatment resulted in both the protection of cytotoxicity and a reduction of proinflammatory cytokines. These results suggest that the autophagy activators can be a new protection regimen for anti-pollution. Therefore, CD extract can be used for anti-inflammatory and anti-pollution cosmetic ingredients.
Collapse
|
32
|
Fasting and Its Impact on Skin Anatomy, Physiology, and Physiopathology: A Comprehensive Review of the Literature. Nutrients 2019; 11:nu11020249. [PMID: 30678053 PMCID: PMC6413166 DOI: 10.3390/nu11020249] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/13/2019] [Accepted: 01/18/2019] [Indexed: 12/14/2022] Open
Abstract
Skin serves as the first protective line and barrier of the body. Like many other organs, skin can be affected by several disorders in response to external factors such as pathogens, ultraviolet light, and pollution, as well as endogenous alterations related to aging and/or oxidative stress disturbance. Researchers have reported new insights into how skin cells are altered in response to caloric restriction diets in mammals. One of the most well-known caloric restriction diets is the Ramadan intermittent fasting, which is a radical change in the diet plan of practitioners for the period of one lunar month. Ramadan fasting represents the fourth of the five pillars of the Islamic creed. Even though infirm individuals are waived to take part in this religious duty, patients with various health problems, including those with different skin disorders, might choose to share this event with peers and family members. No standardized protocols or guidelines exist, however, to advise their physicians on the proper management of their patients' condition during fasting. With an increasing Muslim population living in Western countries, this topic has started to draw substantial attention, not only of Middle-Eastern physicians, but also of clinicians in the West. For this purpose, we carried out a comprehensive overview on the topic. Our main findings are that: (1) there is a strong need for evidence-based suggestions and guidance. Literature on the impact of the Ramadan fasting, as well as of other kinds of fasting, on skin diseases is scarce and of poor quality, as well as the information available from the Internet; (2) patients willing to fast should be advised about the importance of taking proper treatments or consider alternative options including administration of trans-dermal/topical drugs, as they are permitted during daylight hours. Further, non-compliance has important, clinical and economic implications for an effective patient management.
Collapse
|