Abstract
The skin inflammasome is a critical component of the immune system, pivotal not only in responding to acute threats but also in contributing to the chronic inflammation associated with aging. This review provides an in-depth examination of the molecular mechanisms of the skin inflammasome, detailing its role in dermatological conditions like acne, atopic dermatitis, psoriasis, and hyperpigmentation, as well as its impact on systemic aging. We explore how longevity science is now focusing on these pathways to enhance skin resilience and potentially extend human health span. This involves innovative approaches such as drug repurposing, understanding the modulation of skin inflammation, and recognizing the systemic health reflections through the gut-skin axis.
Keywords
Classical immunology, Clinical immunology, Dermatology, Immune cells and organs, Immunomodulation, Immunotherapy, Inflammasome, Longevity, Longevity Science, Skin
Introduction
Inflammation is integral to the skin’s defensive strategy, essential for wound repair and the elimination of pathogens. However, when inflammation becomes dysregulated, it can lead to chronic health issues and accelerate aging. The NLRP3 inflammasome in the skin acts as a sentinel, capable of detecting both external pathogens and internal cellular stress. Longevity science seeks to extend not only lifespan but also health span, defined as the period of life spent in good health, free from chronic diseases and disabilities associated with aging. Central to this endeavor is understanding how aging interacts with inflammasome pathways. With advancing age, the regulation of these pathways becomes impaired, leading to heightened basal activation of the inflammasome. This contributes to persistent low-grade inflammation, known as inflammaging, which accelerates aging processes and predisposes individuals to age-related diseases. This review aims to dissect the intricate connections between the skin inflammasome, aging, and longevity, compiling recent research to propose novel therapeutic strategies.
Inflammaging: The Chronic Inflammation of Aging
The term “inflammaging” describes the chronic, low-grade inflammation that drives aging. This review explores this concept, detailing how inflammasomes facilitate this process. We discuss how skin cells, including keratinocytes and macrophages, harbor inflammasome components, and how these systems transition from beneficial to detrimental as we age:
Inflammaging and aging biomarkers
The link between inflammaging, telomere shortening, epigenetic alterations, and mitochondrial dysfunction has been solidified, indicating that inflammasome activity contributes to systemic aging processes [7,20]. Recent studies suggest that the inflammasome’s role in aging might be mediated by its response to cellular damage and stress, influencing the aging trajectory at a molecular level.
Cellular senescence
Senescent cells contribute significantly to inflammaging by releasing SASP, a cocktail of inflammatory cytokines including IL-1β, which can stimulate nearby inflammasomes, thus perpetuating inflammation and aging [8,26]. This not only affects the local environment of the skin but also has systemic implications.
Nad+ decline
With age, the decline in NAD+ levels is associated with mitochondrial dysfunction, which is a known activator of the NLRP3 inflammasome, thereby linking metabolic health directly to the inflammatory processes of aging [9,27]. This relationship underscores the potential of NAD+ supplementation in aging research.
The Skin Inflammasome: Molecular Mechanisms and Pathways
Nlrp3 inflammasome activation
Priming: The process begins with the upregulation of NLRP3 in response to signals like LPS from bacteria or endogenous danger signals such as ATP or uric acid. The transcription factor NF-κB not only increases NLRP3 expression but also initiates the transcription of pro-IL-1β, preparing the cell for an inflammatory response. This priming phase is crucial for sensitizing cells to subsequent activation signals, a mechanism that parallels arming a defense system [1,2]. Additionally, recent research has shown that other signaling pathways, including those mediated by DAMPs (damage-associated molecular patterns) and PAMPs (pathogen-associated molecular patterns), can also prime the inflammasome [20].
Activation: Following priming, the inflammasome requires a secondary signal, often from cellular stress like ion flux, mitochondrial damage, or lysosomal rupture, to activate. Here, NLRP3 polymerizes with ASC and caspase-1, leading to the cleavage of pro-inflammatory cytokines into their active forms, IL-1β and IL-18, initiating an inflammatory cascade. This dual-signal requirement is evolutionary, ensuring that activation occurs only under appropriate conditions, thus preventing unnecessary inflammation [2,3,21]. The involvement of various ion channels, including the P2X7 receptor for ATP, has been highlighted as key in this activation step [2].
Inflammasome in skin pathology
Acne vulgaris: Propionibacterium acnes in the sebaceous glands activates the NLRP3 inflammasome through its bacterial products like lipases or direct interaction with immune cells. This leads to the formation of inflammatory lesions, indicating the microbiome's critical role in skin health [4,22]. Recent studies have also begun to explore how the skin's resident microbiome might influence inflammasome activity, suggesting potential therapeutic avenues through microbiome modulation.
Atopic dermatitis: Continuous exposure to allergens or irritants results in sustained inflammasome activation, where IL-1β not only drives inflammation but also compromises the skin barrier, perpetuating a cycle of 'itch-scratch'. This cycle not only aggravates barrier function but also promotes further inflammation, illustrating the complex feedback between skin barrier integrity and inflammasome activity [5,23]. The role of IL-33, another inflammasome-related cytokine, has also been implicated in the chronic nature of this condition.
Psoriasis: The NLRP3 inflammasome plays a central role in psoriatic lesions where IL-1β sustains a feedback loop with IL-17 and IL-23, leading to keratinocyte proliferation and plaque formation. The persistent activation of this inflammasome highlights its potential as a therapeutic target, where blocking specific steps in this pathway could alleviate symptoms [6,24]. Recent advances in understanding the genetic basis of psoriasis suggest that certain genetic variants might predispose individuals to exaggerated inflammasome responses.
Hyperpigmentation: While not directly driven by inflammasomes, chronic inflammation can influence melanocytes through cytokines like IL-6, leading to melanin overproduction and pigmentation changes. This indirect link between inflammation and skin pigmentation underscores how inflammatory processes can have cosmetic as well as health implications [7]. The role of inflammasome activation in conditions like melasma is an area of growing interest.
Aging and inflammaging
Increased basal activation: The cumulative effect of UV radiation, oxidative stress, and environmental pollutants over a lifetime leads to a higher baseline of inflammasome activity. This contributes to the concept of 'inflammaging', where persistent low-grade inflammation hastens aging processes, particularly in the skin [8,25]. Recent findings also link this increased activation to the accumulation of cellular debris and senescent cells, further exacerbating the aging process.
Senescent cells: These cells, unable to proliferate, secrete SASP, which includes cytokines like IL-1β that stimulate inflammasome activation in neighboring cells, thus perpetuating inflammation. This phenomenon not only affects local skin aging but has systemic repercussions, suggesting that targeting senescent cells could be beneficial for both skin and overall health [9,26].
Nad+ decline: As NAD+ levels fall with age, this impacts cellular energy and repair mechanisms, leading to mitochondrial dysfunction, which is a significant activator of the NLRP3 inflammasome. This connection between metabolic health and inflammation underscores the importance of metabolic interventions in aging research [10,27]. The potential of NAD+ precursors like NMN to reverse these effects is under active investigation.
Longevity Science and Skin Health
Nad+ and sirtuins
Nad+ boosting: Therapies aimed at increasing NAD+ levels, such as NMN or NR supplementation, are being explored for their potential to enhance DNA repair, improve mitochondrial function, and thereby reduce inflammasome activity, offering a novel approach to aging [10]. Clinical trials are underway to translate these preclinical benefits to human health.
Sirtuins: SIRT1, an NAD+-dependent protein, has shown anti-inflammatory effects that could mimic the longevity benefits of caloric restriction. Its ability to modulate inflammatory pathways might provide a pathway to reduce skin aging and inflammation [11,27]. Recent studies have also linked SIRT1 to the regulation of skin's immune responses, suggesting broader implications for skin health.
Senolytics
Dasatinib and quercetin: These compounds selectively eliminate senescent cells, reducing the secretion of SASP and thus chronic inflammation. This strategy is being investigated for managing age-related skin conditions, potentially offering a new paradigm in dermatological treatment [12,28]. The combination's efficacy in animal models has been promising, though human trials are necessary to confirm these benefits.
Targeting SASP: Rather than eliminating senescent cells, research is also focusing on modulating SASP to control inflammation without the potential risks of broad immune suppression. This could be particularly beneficial for aging skin, promoting a healthier aging process [13].
Other longevity molecules
AMPK activators: Metformin, traditionally used for diabetes, has shown potential in inhibiting inflammasome assembly, linking metabolic health directly to inflammation control in aging. Its pleiotropic effects might extend beyond diabetes management to include anti-aging properties for the skin [14,29].
mTOR inhibitors: Rapamycin, known for its immunosuppressive properties, can modulate mTOR pathways, which are implicated in aging and inflammation. By lowering inflammatory responses, it might slow down aging processes, including those affecting the skin [15]. The use of rapamycin in topical formulations is thus being explored.
Longevity Science and the Gut-skin Axis
Microbiome influence
The gut microbiome’s balance significantly influences systemic inflammation, which in turn can affect skin health. Dysbiosis leads to increased gut permeability, allowing LPS and other microbial products to activate skin inflammasomes, suggesting that maintaining gut health might indirectly benefit the skin [18,32].
Leaky gut and systemic inflammation
Increased intestinal permeability results in systemic inflammation, which can activate skin inflammasomes, contributing to conditions like psoriasis or general skin aging. This relationship supports the use of prebiotics, probiotics, or dietary interventions to manage skin conditions through gut health [19].
Psycho-neuro-endocrine pathways
The gut influences systemic stress responses and hormone levels, which can exacerbate skin conditions by promoting inflammation or collagen degradation. Cortisol, for instance, from chronic stress, can compromise skin barrier function and immune response, accelerating skin aging [5,6,30].
Dietary influence
Diets rich in anti-inflammatory nutrients like omega-3 fatty acids, antioxidants, and fiber can support a balanced gut microbiome, potentially reducing systemic inflammation and benefiting skin health [7,31]. The Mediterranean diet, in particular, has been associated with lower levels of inflammatory markers.
Immunological linkage
The gut educates the immune system, which can modulate skin immunity. Dysbiosis can lead to an imbalance in cytokine production, exacerbating inflammatory skin conditions. This suggests that interventions aimed at restoring gut microbial balance could have therapeutic implications for skin health [9,10,32].
Specific evidence
Recent studies provide concrete evidence for the gut-skin axis. For example, a randomized controlled trial showed that supplementation with Lactobacillus rhamnosus improved skin barrier function and reduced inflammation in patients with atopic dermatitis [42]. Additionally, dietary interventions like the Mediterranean diet, rich in omega-3 fatty acids and antioxidants, have been linked to reduced systemic inflammation and improved skin health [43]. Regular physical exercise has also been shown to modulate immune responses and reduce inflammasome activation, potentially benefiting skin health [44].
Drug Repurposing and Systemic Health Implications
Metformin
Its anti-inflammatory properties are now being explored for dermatological benefits, potentially modulating inflammasome activity in the skin. This could offer dual benefits in managing both metabolic disorders and signs of skin aging [16,33]. The Targeting Aging with Metformin (TAME) trial is investigating metformin’s potential to delay age-related diseases, including those affecting the skin, providing a robust framework for its application in aging research [14].
Statins
Beyond cholesterol management, statins have anti-inflammatory effects that could inhibit inflammasome activation, providing a novel approach to treat inflammatory skin diseases [17]. Studies suggest statins may benefit conditions like psoriasis due to their anti-inflammatory properties, with ongoing research exploring their efficacy in dermatology [45]. Their role in skin health is still under investigation but shows promise.
Therapeutic Strategies
The dual role of the inflammasome in health and disease has led to the development of targeted therapies, such as biologics that block cytokines like IL-1β in psoriasis, demonstrating how molecular insights into inflammasome biology can guide treatment [6,34].
Despite the promise of senolytics and NAD+ boosters, their use in dermatology faces challenges regarding safety, cost, and efficacy in human trials. It is crucial to temper expectations with the current state of research, highlighting the need for more comprehensive studies to validate these approaches [10,26].
Challenges and considerations
Translating these findings into clinical practice involves hurdles such as ensuring long-term safety and sustainability. Senolytics, while effective in preclinical models, may have off-target effects on healthy cells, and their long-term impact on tissue regeneration requires evaluation. NAD+ boosters need further study to determine optimal dosing and potential side effects. Drug repurposing and lifestyle interventions, though promising, face challenges like individual variability and adherence, necessitating robust clinical trials to establish their efficacy and safety in real-world settings.
Other Contributors to Skin Aging and Inflammation
Oxidative stress
Chronic exposure to ROS from environmental or internal sources leads to cellular damage, promoting inflammation and aging. While antioxidants can mitigate some damage, their direct impact on modulating inflammasome activity is still under investigation [7,35].
Mitochondrial dysfunction
Mitochondria’s role in energy production and inflammasome activation means that their dysfunction with age can increase ROS, triggering NLRP3 inflammasome activation, directly linking metabolic health to skin aging [8,36].
External factors
UV radiation and pollution not only damage DNA but also disrupts the skin barrier, allowing for increased inflammasome activation by environmental agents. Research into protective mechanisms against these factors is vital for preventing skin aging and inflammation [7,37].
Actionable Insights for Translational Research
Personalized therapies
The future of dermatological treatment lies in personalizing strategies based on individual inflammatory profiles, genetic predispositions, and lifestyle factors. This approach could optimize treatment outcomes while minimizing side effects [38]. However, practical implementation faces challenges such as the high cost of genetic profiling and the need for large-scale validation studies.
Systemic interventions
Exploring how systemic health interventions like diet, exercise, or microbiome modulation can influence skin inflammasome activity might lead to innovative therapeutic strategies [39].
Topical treatments
There is a demand for less invasive, more effective topical agents that can target inflammasome pathways directly. Advances in drug delivery systems, like nanoparticles, could revolutionize this field [40].
Biomarkers and monitoring
Identifying reliable biomarkers for inflammasome activity in the skin would enhance our ability to monitor treatment efficacy and disease progression, bridging the gap between research and clinical application [41].
Conclusion
The skin inflammasome plays a pivotal role in both protective and pathological inflammation, making it a critical target for interventions aimed at extending health span. This review has synthesized current insights into how inflammasomes contribute to aging and skin disorders, highlighting the innovative approaches of longevity science to mitigate inflammation. The gut-skin axis underscores the systemic nature of health, suggesting that interventions in one area might yield benefits elsewhere. By leveraging drug repurposing, personalized medicine, and lifestyle modifications, we can better manage aging. This review not only links dermatology with longevity science but also opens new research and therapeutic avenues, potentially reshaping our approach to aging and health maintenance. However, limitations exist, such as reliance on preclinical models and the need for more human studies to validate findings. Future research should address specific questions, such as the role of gut microbiota in skin inflammasome activation, the development of topical senolytics, and the integration of systemic and dermatological interventions.
References
2. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002 Aug;10(2):417-26.
3. Qin M, Pirouz A, Kim MH, Krutzik SR, Garbán HJ, Kim J. Propionibacterium acnes Induces IL-1? secretion via the NLRP3 inflammasome in human monocytes. J Invest Dermatol. 2014 Feb;134(2):381-8.
4. Dinarello CA, et al. The role of inflammasomes in inflammatory skin disorders. Journal of Investigative Dermatology. 2013;133(7):1668-73.
5. Fujita H, et al. Contributions of IL-1? and IL-18 to psoriasis pathogenesis. Journal of Dermatological Science. 2011;63(2):118-23.
6. Kim M, Park J, Park H, et al. The Role of Inflammatory Mediators in Melanogenesis. International Journal of Molecular Sciences. 2018;19(3):840.
7. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014 Jun;69 Suppl 1:S4-9.
8. Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99-118.
9. Imai S, Guarente L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014 Aug;24(8):464-71.
10. Yoshino J, Baur JA, Imai SI. NAD+ Intermediates: The Biology and Therapeutic Potential of NMN and NR. Cell Metab. 2018 Mar 6;27(3):513-28.
11. Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. 2006 Jun;5(6):493-506.
12. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015 Aug;14(4):644-58.
13. Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The Clinical Potential of Senolytic Drugs. J Am Geriatr Soc. 2017 Oct;65(10):2297-301.
14. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a Tool to Target Aging. Cell Metab. 2016 Jun 14;23(6):1060-5.
15. Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013 Jan 17;493(7432):338-45.
16. Prabhakar O, Thakur A. Overview of metformin: Special emphasis on its anti-inflammatory properties. Current Diabetes Reviews. 2017;13(6):551-60.
17. Hennessy E, Scholz CC, O’Brien PJ. The role of PPARs in the anti-inflammatory effects of statins. PPAR Research. 2016;2016:9874528.
18. Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol. 2018 Jul 10;9:1459.
19. Bischoff SC, Barbara G, Buurman W, Ockhuizen T, Schulzke JD, Serino M, et al. Intestinal permeability--a new target for disease prevention and therapy. BMC Gastroenterol. 2014 Nov 18;14:189.
20. Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol. 2016 Mar;13(2):148-59.
21. Schroder K, Tschopp J. The inflammasomes. Cell. 2010 Mar 19;140(6):821-32.
22. Dreno B, Martin R, Moyal D, Henley JB, Khammari A, Seité S. Skin microbiome and acne vulgaris: Staphylococcus, Propionibacterium, and beyond. British Journal of Dermatology. 2017;177(2):353-60.
23. Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med. 2017 Feb 22;9(378):eaah4680.
24. Mabuchi T, Chang TW. Psoriasis and Its Comorbidities. Journal of Dermatology. 2018;45(2):134-43.
25. Chen JH, Dyer DC. Advanced glycation end-products (AGEs) and their receptor (RAGE) in skin aging and disease. International Journal of Molecular Sciences. 2019;20(1):105.
26. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018 Aug;24(8):1246-56.
27. Mostoslavsky R, Chua KF, Lombard DB, Pang WW, Fischer MR, Gellon L, et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell. 2006 Jan 27;124(2):315-29.
28. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018 Jun;28(6):436-53.
29. Blagosklonny MV. Calorie restriction: decelerating mTOR-driven aging from cells to organisms (including humans). Cell Cycle. 2010 Feb 15;9(4):683-8.
30. Arck PC, Handjiski B, Hagen E, Joachim R, Klapp BF, Paus R. Indications for a 'brain-hair follicle axis (BHA)': inhibition of keratinocyte proliferation and up-regulation of keratinocyte apoptosis in telogen hair follicles by stress and substance P. FASEB J. 2001 Nov;15(13):2536-8.
31. Guasch-Ferré M, Merino J, Sun Q, Fitó M, Salas-Salvadó J. Dietary Polyphenols, Mediterranean Diet, Prediabetes, and Type 2 Diabetes: A Narrative Review of the Evidence. Oxid Med Cell Longev. 2017;2017:6723931.
32. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014 Mar 27;157(1):121-41.
33. Del Barco S, Vazquez-Martin A, Cufí S, Oliveras-Ferraros C, Bosch-Barrera J, Joven J, et al. Metformin: multi-faceted protection against cancer. Oncotarget. 2011 Dec;2(12):896-917.
34. Menter A, Strober BE, Kaplan DH, Kivelevitch D, Prater EF, Stoff B, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019 Apr;80(4):1029-72.
35. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 2007;39(1):44-84.
36. Zinovkin RA, Zamyatnin AA. Mitochondria-targeted drugs. Current Molecular Pharmacology. 2019 Aug 1;12(3):202-14.
37. Krutmann J, Bouloc A, Sore G, Bernard BA, Passeron T. The skin aging exposome. J Dermatol Sci. 2017 Mar;85(3):152-161.
38. Potaczek DP, Garn H, Unger SD. Inflammasomes in atopic dermatitis and contact allergy. Current Opinion in Allergy and Clinical Immunology. 2011;11(6):617-23.
39. Longo VD, Panda S. Fasting, Circadian Rhythms, and Time-Restricted Feeding in Healthy Lifespan. Cell Metab. 2016 Jun 14;23(6):1048-59.
40. Lboutounne, H., Faivre, V., Faison, F., et al. (2004). Characterization of transport of cholecalciferol (vitamin D3) in liposomes through stratum corneum using steady-state diffusion and confocal Raman spectroscopy. Journal of Controlled Release, 99(3), 345-53.
41. Youn JI, Park JY, Jo SJ, et al. In vivo monitoring of skin inflammasome activity in psoriasis by near-infrared fluorescence imaging. Journal of Investigative Dermatology. 2019;139(4):923-32.
42. Guéniche A, et al. Probiotics for skin benefits. Journal of Applied Microbiology. 2010;109(4):1155-61.
43. Cosgrove MC, Franco OH, Granger SP, Murray PG, Mayes AE. Dietary nutrient intakes and skin-aging appearance among middle-aged American women. Am J Clin Nutr. 2007 Oct;86(4):1225-31.
44. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo MA. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol. 2011 Aug 5;11(9):607-15.
45. Namazi MR. Statins: novel additions to the dermatologic arsenal? Exp Dermatol. 2004 Jun;13(6):337-9.