Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence
Abstract
:1. Introduction
2. Gut Microbiome and Human Aging
3. Cellular Senescence as a Central Dogma of Human Aging
Inducers and Metabolic Activity of Senescent Cells
4. Gut Microbiome and Its Link to Cellular Senescence
4.1. Microbial Dysbiosis May Influence a Senescent State
4.2. Microbial Metabolites Promote SASP Damage
5. Skin Health and Its Relation to Microbiome and Aging
5.1. Skin Homeostasis Plays a Role in Healthy Aging
5.2. Gut-Skin Axis Connects Skin Dysfunction to Metabolic Disruption
5.3. Age-Related Skin Microbial Dysbiosis
6. The Impact of Microbiome and Cellular Senescence on Skin Health and Diseases
6.1. Consequences of Senescent Cell Accumulation in the Skin
6.2. Skin Diseases Associated with Senescence and Microbial Implications
6.2.1. Skin Health and Aging
6.2.2. Skin Carcinogenesis
6.2.3. Dyspigmentation
6.2.4. Psoriasis and Atopic Dermatitis
6.2.5. Acne Vulgaris
6.2.6. Chronic Wounds
6.2.7. Skin Immunity
7. Therapeutic Strategies for Targeting Cellular Senescence
7.1. Pharmaceutical Agents and Their Drawbacks
7.1.1. Senolytics
7.1.2. Senostatics
7.2. Nutraceuticals and Diet as a Novel Approach to Fighting Senescence
7.3. Probiotics and Prebiotics as New Therapeutics for Managing Aging
8. Use of Metabolomics in Targeting Senescence
9. Healthy Aging and Skin Health Product Development Opportunities
10. Future Implications
11. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Das, B.; Nair, G.B. Homeostasis and dysbiosis of the gut microbiome in health and disease. J. Biosci. 2019, 44, 117. [Google Scholar] [CrossRef] [PubMed]
- Belizário, J.E.; Faintuch, J.; Garay-Malpartida, M. Gut Microbiome Dysbiosis and Immunometabolism: New Frontiers for Treatment of Metabolic Diseases. Mediat. Inflamm. 2018, 2018, 2037838. [Google Scholar] [CrossRef]
- Srivastava, S.; Singh, A.; Sandeep, K.; Yadav, D. Epigenetic Regulation of Gut Microbial Dysbiosis. Indian J. Microbiol. 2021, 61, 125–129. [Google Scholar] [CrossRef]
- Thevaranjan, N.; Puchta, A.; Schulz, C.; Naidoo, A.; Szamosi, J.C.; Verschoor, C.P.; Loukov, D.; Schenck, L.P.; Jury, J.; Foley, K.P.; et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe 2017, 21, 455–466.e4. [Google Scholar] [CrossRef] [Green Version]
- Vaiserman, A.; Romanenko, M.; Piven, L.; Moseiko, V.; Lushchak, O.; Kryzhanovska, N.; Guryanov, V.; Koliada, A. Differences in the gut Firmicutes to Bacteroidetes ratio across age groups in healthy Ukrainian population. BMC Microbiol. 2020, 20, 221. [Google Scholar] [CrossRef]
- Fernandes, J.; Su, W.; Rahat-Rozenbloom, S.; Wolever, T.M.; Comelli, E.M. Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr. Diabetes 2014, 4, e121. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.-S.; Choi, C.W.; Shin, H.; Jin, A.-P.; Bae, J.-S.; Han, M.; Seo, E.Y.; Chun, J.; Chung, J.H. Comparison of the Gut Microbiota of Centenarians in Longevity Villages of South Korea with Those of Other Age Groups. J. Microbiol. Biotechnol. 2019, 29, 429–440. [Google Scholar] [CrossRef] [PubMed]
- Kong, F.; Hua, Y.; Zeng, B.; Ning, R.; Li, Y.; Zhao, J. Gut microbiota signatures of longevity. Curr. Biol. 2016, 26, R832–R833. [Google Scholar] [CrossRef] [Green Version]
- Franceschi, C.; Capri, M.; Monti, D.; Giunta, S.; Olivieri, F.; Sevini, F.; Panourgia, M.P.; Invidia, L.; Celani, L.; Scurti, M.; et al. Inflammaging and anti-inflammaging: A systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 2007, 128, 92–105. [Google Scholar] [CrossRef]
- Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
- Borghesan, M.; Hoogaars, W.M.H.; Varela-Eirin, M.; Talma, N.; Demaria, M. A Senescence-Centric View of Aging: Implications for Longevity and Disease. Trends Cell Biol. 2020, 30, 777–791. [Google Scholar] [CrossRef] [PubMed]
- Wang, A.S.; Dreesen, O. Biomarkers of Cellular Senescence and Skin Aging. Front. Genet. 2018, 9, 247:1–247:14. [Google Scholar] [CrossRef]
- Gonzalez-Meljem, J.M.; Apps, J.R.; Fraser, H.C.; Martinez-Barbera, J.P. Paracrine roles of cellular senescence in promoting tumourigenesis. Br. J. Cancer 2018, 118, 1283–1288. [Google Scholar] [CrossRef]
- Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
- Kirkland, J.L.; Tchkonia, T. Senolytic drugs: From discovery to translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef]
- Nakagami, H. Cellular senescence and senescence-associated T cells as a potential therapeutic target. Geriatr. Gerontol. Int. 2020, 20, 97–100. [Google Scholar] [CrossRef] [Green Version]
- Kale, S.L.; Gorry, M.; Ray, P.; Ray, A. Cellular senescence and SASP in the pathobiology of IFN-γ severe asthma. J. Immunol. 2020, 204, 16. [Google Scholar]
- Mas-Bargues, C.; Borras, C.; Vina, J. Bcl-xL as a Modulator of Senescence and Aging. Int. J. Mol. Sci. 2021, 22, 1527. [Google Scholar] [CrossRef] [PubMed]
- Rochette, P.J.; Brash, D.E. Progressive apoptosis resistance prior to senescence and control by the anti-apoptotic protein BCL-xL. Mech. Ageing Dev. 2008, 129, 207–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jonchère, B.; Vétillard, A.; Toutain, B.; Lam, D.; Bernard, A.C.; Henry, C.; De Carné Trécesson, S.; Gamelin, E.; Juin, P.; Guette, C.; et al. Irinotecan treatment and senescence failure promote the emergence of more transformed and invasive cells that depend on anti-apoptotic Mcl-1. Oncotarget 2015, 6, 409–426. [Google Scholar] [CrossRef] [Green Version]
- Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O.; et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [Green Version]
- Stein, G.H.; Drullinger, L.F.; Soulard, A.; Dulić, V. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol. Cell Biol. 1999, 19, 2109–2117. [Google Scholar] [CrossRef] [Green Version]
- Kuilman, T.; Michaloglou, C.; Mooi, W.J.; Peeper, D.S. The essence of senescence. Genes Dev. 2010, 24, 2463–2479. [Google Scholar] [CrossRef] [Green Version]
- Demaria, M.; Desprez, P.Y.; Campisi, J.; Velarde, M.C. Cell Autonomous and Non-Autonomous Effects of Senescent Cells in the Skin. J. Investig. Dermatol. 2015, 135, 1722–1726. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Wan, M. Chapter Three—A tale of the good and bad: Cell senescence in bone homeostasis and disease. In International Review of Cell and Molecular Biology; Galluzzi, L., Ed.; Academic Press: Cambridge, MA, USA, 2019; Volume 346, pp. 97–128. [Google Scholar]
- Attaallah, A.; Lenzi, M.; Marchionni, S.; Bincoletto, G.; Cocchi, V.; Croco, E.; Hrelia, P.; Hrelia, S.; Sell, C.; Lorenzini, A. A pro longevity role for cellular senescence. GeroScience 2020, 42, 867–879. [Google Scholar] [CrossRef]
- Victorelli, S.; Lagnado, A.; Halim, J.; Moore, W.; Talbot, D.; Barrett, K.; Chapman, J.; Birch, J.; Ogrodnik, M.; Meves, A.; et al. Senescent human melanocytes drive skin ageing via paracrine telomere dysfunction. EMBO J. 2019, 38, e101982. [Google Scholar] [CrossRef]
- Fitsiou, E.; Pulido, T.; Campisi, J.; Alimirah, F.; Demaria, M. Cellular Senescence and the Senescence-Associated Secretory Phenotype as Drivers of Skin Photoaging. J. Investig. Dermatol. 2021, 141, 1119–1126. [Google Scholar] [CrossRef]
- Wiley, C.D.; Campisi, J. The metabolic roots of senescence: Mechanisms and opportunities for intervention. Nature 2021, 3, 1290–1301. [Google Scholar] [CrossRef]
- Rovillain, E.; Mansfield, L.; Caetano, C.; Alvarez-Fernandez, M.; Caballero, O.L.; Medema, R.H.; Hummerich, H.; Jat, P.S. Activation of nuclear factor-kappa B signalling promotes cellular senescence. Oncogene 2011, 30, 2356–2366. [Google Scholar] [CrossRef] [Green Version]
- Yoshino, J.; Baur, J.A.; Imai, S.I. NAD(+) Intermediates: The Biology and Therapeutic Potential of NMN and NR. Cell Metab. 2018, 27, 513–528. [Google Scholar] [CrossRef] [Green Version]
- Sabbatinelli, J.; Prattichizzo, F.; Olivieri, F.; Procopio, A.D.; Rippo, M.R.; Giuliani, A. Where Metabolism Meets Senescence: Focus on Endothelial Cells. Front. Physiol. 2019, 10, 1523. [Google Scholar] [CrossRef] [Green Version]
- Maceyka, M.; Spiegel, S. Sphingolipid metabolites in inflammatory disease. Nature 2014, 510, 58–67. [Google Scholar] [CrossRef] [Green Version]
- Wiley, C.D.; Sharma, R.; Davis, S.S.; Lopez-Dominguez, J.A.; Mitchell, K.P.; Wiley, S.; Alimirah, F.; Kim, D.E.; Payne, T.; Rosko, A.; et al. Oxylipin biosynthesis reinforces cellular senescence and allows detection of senolysis. Cell Metab. 2021, 33, 1124–1136.e5. [Google Scholar] [CrossRef]
- Metchnikoff, E. The Prolongation of Life; G.P. Putnam’s Sons: New York, NY, USA, 1908. [Google Scholar]
- Saccon, T.D.; Nagpal, R.; Yadav, H.; Cavalcante, M.B.; Nunes, A.D.d.C.; Schneider, A.; Gesing, A.; Hughes, B.; Yousefzadeh, M.; Tchkonia, T.; et al. Senolytic Combination of Dasatinib and Quercetin Alleviates Intestinal Senescence and Inflammation and Modulates the Gut Microbiome in Aged Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 1895–1905. [Google Scholar] [CrossRef]
- Krautkramer, K.A.; Fan, J.; Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 2021, 19, 77–94. [Google Scholar] [CrossRef] [PubMed]
- Szántó, M.; Dózsa, A.; Antal, D.; Szabó, K.; Kemény, L.; Bai, P. Targeting the gut-skin axis—Probiotics as new tools for skin disorder management? Exp. Dermatol. 2019, 28, 1210–1218. [Google Scholar] [CrossRef] [Green Version]
- Sivaprakasam, S.; Prasad, P.D.; Singh, N. Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol. Ther. 2016, 164, 144–151. [Google Scholar] [CrossRef] [Green Version]
- Matt, S.M.; Allen, J.M.; Lawson, M.A.; Mailing, L.J.; Woods, J.A.; Johnson, R.W. Butyrate and Dietary Soluble Fiber Improve Neuroinflammation Associated with Aging in Mice. Front. Immunol. 2018, 9, 1832. [Google Scholar] [CrossRef]
- Ajouz, H.; Mukherji, D.; Shamseddine, A. Secondary bile acids: An underrecognized cause of colon cancer. World J. Surg. Oncol. 2014, 12, 164. [Google Scholar] [CrossRef] [Green Version]
- Heinken, A.; Ravcheev, D.A.; Baldini, F.; Heirendt, L.; Fleming, R.M.T.; Thiele, I. Systematic assessment of secondary bile acid metabolism in gut microbes reveals distinct metabolic capabilities in inflammatory bowel disease. Microbiome 2019, 7, 75. [Google Scholar] [CrossRef] [PubMed]
- Biragyn, A.; Ferrucci, L. Gut dysbiosis: A potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol. 2018, 19, e295–e304. [Google Scholar] [CrossRef]
- Lee, K.; Kim, H.J.; Kim, S.A.; Park, S.-D.; Shim, J.-J.; Lee, J.-L. Exopolysaccharide from Lactobacillus plantarum HY7714 Protects against Skin Aging through Skin–Gut Axis Communication. Molecules 2021, 26, 1651. [Google Scholar] [CrossRef] [PubMed]
- Coates, M.; Lee, M.J.; Norton, D.; MacLeod, A.S. The Skin and Intestinal Microbiota and Their Specific Innate Immune Systems. Front. Immunol. 2019, 10, 2950. [Google Scholar] [CrossRef] [PubMed]
- Parrado, C.; Mercado-Saenz, S.; Perez-Davo, A.; Gilaberte, Y.; Gonzalez, S.; Juarranz, A. Environmental Stressors on Skin Aging. Mechanistic Insights. Front. Pharmacol. 2019, 10, 759. [Google Scholar] [CrossRef] [PubMed]
- Hu, L.; Mauro, T.M.; Dang, E.; Man, G.; Zhang, J.; Lee, D.; Wang, G.; Feingold, K.R.; Elias, P.M.; Man, M.-Q. Epidermal Dysfunction Leads to an Age-Associated Increase in Levels of Serum Inflammatory Cytokines. J. Investig. Dermatol. 2017, 137, 1277–1285. [Google Scholar] [CrossRef] [Green Version]
- Ye, L.; Mauro, T.M.; Dang, E.; Wang, G.; Hu, L.Z.; Yu, C.; Jeong, S.; Feingold, K.; Elias, P.M.; Lv, C.Z.; et al. Topical applications of an emollient reduce circulating pro-inflammatory cytokine levels in chronically aged humans: A pilot clinical study. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 2197–2201. [Google Scholar] [CrossRef]
- De Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut–Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef] [PubMed]
- Miyazaki, K.; Masuoka, N.; Kano, M.; Iizuka, R. Bifidobacterium fermented milk and galacto-oligosaccharides lead to improved skin health by decreasing phenols production by gut microbiota. Benef. Microbes 2014, 5, 121–128. [Google Scholar] [CrossRef]
- Salem, I.; Ramser, A.; Isham, N.; Ghannoum, M.A. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front. Microbiol. 2018, 9, 1459. [Google Scholar] [CrossRef] [Green Version]
- Steele, C.E.; Morrell, D.; Evans, M. Metabolic syndrome and inflammatory skin conditions. Curr. Opin. Pediatr. 2019, 31, 515–522. [Google Scholar] [CrossRef]
- Chen, L.; Li, J.; Zhu, W.; Kuang, Y.; Liu, T.; Zhang, W.; Chen, X.; Peng, C. Skin and Gut Microbiome in Psoriasis: Gaining Insight into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front. Microbiol. 2020, 11, 3201. [Google Scholar] [CrossRef]
- Abuabara, K.; Azfar, R.S.; Shin, D.B.; Neimann, A.L.; Troxel, A.B.; Gelfand, J.M. Cause-specific mortality in patients with severe psoriasis: A population-based cohort study in the U.K. Br. J. Dermatol. 2010, 163, 586–592. [Google Scholar] [CrossRef] [Green Version]
- Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.-J.; Kim, J.J.; Myeong, N.R.; Kim, T.; Kim, D.; An, S.; Kim, H.; Park, T.; Jang, S.I.; Yeon, J.H.; et al. Segregation of age-related skin microbiome characteristics by functionality. Sci. Rep. 2019, 9, 16748. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Bai, X.; Peng, T.; Yi, X.; Luo, L.; Yang, J.; Liu, J.; Wang, Y.; He, T.; Wang, X.; et al. New Insights into the Skin Microbial Communities and Skin Aging. Front. Microbiol. 2020, 11, 2603. [Google Scholar] [CrossRef] [PubMed]
- Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef] [Green Version]
- Wyld, L.; Bellantuono, I.; Tchkonia, T.; Morgan, J.; Turner, O.; Foss, F.; George, J.; Danson, S.; Kirkland, J.L. Senescence and Cancer: A Review of Clinical Implications of Senescence and Senotherapies. Cancers 2020, 12, 2134. [Google Scholar] [CrossRef]
- Dodig, S.; Čepelak, I.; Pavić, I. Hallmarks of senescence and aging. Biochem. Med. 2019, 29, 030501. [Google Scholar] [CrossRef]
- Debacq-Chainiaux, F.; Leduc, C.; Verbeke, A.; Toussaint, O. UV, stress and aging. Dermato-endocrinology 2012, 4, 236–240. [Google Scholar] [CrossRef] [Green Version]
- Hirobe, T. Keratinocytes regulate the function of melanocytes. Dermatol. Sin. 2014, 32, 200–204. [Google Scholar] [CrossRef] [Green Version]
- Scieglinska, D.; Krawczyk, Z.; Sojka, D.R.; Gogler-Piglowska, A. Heat shock proteins in the physiology and pathophysiology of epidermal keratinocytes. Cell Stress Chaperones 2019, 24, 1027–1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.I.; Choi, S.; Roh, W.S.; Lee, J.H.; Kim, T.-G. Cellular Senescence and Inflammaging in the Skin Microenvironment. Int. J. Mol. Sci. 2021, 22, 3849. [Google Scholar] [CrossRef]
- Velarde, M.C.; Demaria, M. Targeting Senescent Cells: Possible Implications for Delaying Skin Aging: A Mini-Review. Gerontology 2016, 62, 513–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, A.S.; Ong, P.F.; Chojnowski, A.; Clavel, C.; Dreesen, O. Loss of lamin B1 is a biomarker to quantify cellular senescence in photoaged skin. Sci. Rep. 2017, 7, 15678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pilkington, S.M.; Bulfone-Paus, S.; Griffiths, C.E.M.; Watson, R.E.B. Inflammaging and the Skin. J. Investig. Dermatol. 2020, 141, 1087–1095. [Google Scholar] [CrossRef]
- Saez-Atienzar, S.; Masliah, E. Cellular senescence and Alzheimer disease: The egg and the chicken scenario. Nat. Rev. Neurosci. 2020, 21, 433–444. [Google Scholar] [CrossRef]
- Nelson, G.; Kucheryavenko, O.; Wordsworth, J.; von Zglinicki, T. The senescent bystander effect is caused by ROS-activated NF-κB signalling. Mech. Ageing Dev. 2018, 170, 30–36. [Google Scholar] [CrossRef]
- Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef]
- da Silva, P.F.L.; Ogrodnik, M.; Kucheryavenko, O.; Glibert, J.; Miwa, S.; Cameron, K.; Ishaq, A.; Saretzki, G.; Nagaraja-Grellscheid, S.; Nelson, G.; et al. The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell 2019, 18, e12848. [Google Scholar] [CrossRef]
- Shuster, S.; Black, M.M.; McVitie, E. The influence of age and sex on skin thickness, skin collagen and density. Br. J. Dermatol. 1975, 93, 639–643. [Google Scholar] [CrossRef]
- Russell-Goldman, E.; Murphy, G.F. The Pathobiology of Skin Aging: New Insights into an Old Dilemma. Am. J. Pathol. 2020, 190, 1356–1369. [Google Scholar] [CrossRef]
- Zhang, S.; Duan, E. Fighting against Skin Aging: The Way from Bench to Bedside. Cell Transpl. 2018, 27, 729–738. [Google Scholar] [CrossRef]
- Maisel, A.; Waldman, A.; Furlan, K.; Weil, A.; Sacotte, K.; Lazaroff, J.M.; Lin, K.; Aranzazu, D.; Avram, M.M.; Bell, A.; et al. Self-reported Patient Motivations for Seeking Cosmetic Procedures. JAMA Dermatol 2018, 154, 1167–1174. [Google Scholar] [CrossRef]
- Ghosh, K.; Capell, B.C. The Senescence-Associated Secretory Phenotype: Critical Effector in Skin Cancer and Aging. J. Investig. Dermatol. 2016, 136, 2133–2139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, D.E.; Dollé, M.E.T.; Vermeij, W.P.; Gyenis, A.; Vogel, K.; Hoeijmakers, J.H.J.; Wiley, C.D.; Davalos, A.R.; Hasty, P.; Desprez, P.-Y.; et al. Deficiency in the DNA repair protein ERCC1 triggers a link between senescence and apoptosis in human fibroblasts and mouse skin. Aging Cell 2020, 19, e13072. [Google Scholar] [CrossRef] [PubMed]
- Pittayapruek, P.; Meephansan, J.; Prapapan, O.; Komine, M.; Ohtsuki, M. Role of Matrix Metalloproteinases in Photoaging and Photocarcinogenesis. Int. J. Mol. Sci. 2016, 17, 868. [Google Scholar] [CrossRef] [Green Version]
- Philips, N.; Auler, S.; Hugo, R.; Gonzalez, S. Beneficial regulation of matrix metalloproteinases for skin health. Enzym. Res. 2011, 2011, 427285. [Google Scholar] [CrossRef] [Green Version]
- Jabłońska-Trypuć, A.; Matejczyk, M.; Rosochacki, S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J. Enzym. Inhib. Med. Chem. 2016, 31, 177–183. [Google Scholar] [CrossRef] [Green Version]
- Byun, H.-O.; Lee, Y.-K.; Kim, J.-M.; Yoon, G. From cell senescence to age-related diseases: Differential mechanisms of action of senescence-associated secretory phenotypes. BMB Rep. 2015, 48, 549–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Man, M.-Q.; Elias, P.M. Could Inflammaging and Its Sequelae Be Prevented or Mitigated? Clin. Interv. Aging 2019, 14, 2301–2304. [Google Scholar] [CrossRef] [Green Version]
- Vaiserman, A.M.; Koliada, A.K.; Marotta, F. Gut microbiota: A player in aging and a target for anti-aging intervention. Ageing Res. Rev. 2017, 35, 36–45. [Google Scholar] [CrossRef]
- Sharma, D.; Kober, M.M.; Bowe, W.P. Anti-Aging Effects of Probiotics. J. Drugs Dermatol. 2016, 15, 9–12. [Google Scholar]
- Chambers, E.S.; Vukmanovic-Stejic, M. Skin barrier immunity and ageing. Immunology 2020, 160, 116–125. [Google Scholar] [CrossRef] [Green Version]
- Alimirah, F.; Pulido, T.; Valdovinos, A.; Alptekin, S.; Chang, E.; Jones, E.; Diaz, D.A.; Flores, J.; Velarde, M.C.; Demaria, M.; et al. Cellular Senescence Promotes Skin Carcinogenesis through p38MAPK and p44/42MAPK Signaling. Cancer Res. 2020, 80, 3606–3619. [Google Scholar] [CrossRef]
- Coppé, J.-P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.-Y.; Campisi, J. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor. PLoS Biol. 2008, 6, e301. [Google Scholar] [CrossRef] [PubMed]
- Azazmeh, N.; Assouline, B.; Winter, E.; Ruppo, S.; Nevo, Y.; Maly, A.; Meir, K.; Witkiewicz, A.K.; Cohen, J.; Rizou, S.V.; et al. Chronic expression of p16INK4a in the epidermis induces Wnt-mediated hyperplasia and promotes tumor initiation. Nat. Commun. 2020, 11, 2711. [Google Scholar] [CrossRef] [PubMed]
- Liu, D.; Hornsby, P.J. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007, 67, 3117–3126. [Google Scholar] [CrossRef] [Green Version]
- Sherwani, M.A.; Tufail, S.; Muzaffar, A.F.; Yusuf, N. The skin microbiome and immune system: Potential target for chemoprevention? Photodermatol. Photoimmunol. Photomed. 2018, 34, 25–34. [Google Scholar] [CrossRef] [Green Version]
- Fu, C.; Chen, J.; Lu, J.; Yi, L.; Tong, X.; Kang, L.; Pei, S.; Ouyang, Y.; Jiang, L.; Ding, Y.; et al. Roles of inflammation factors in melanogenesis (Review). Mol. Med. Rep. 2020, 21, 1421–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoon, J.E.; Kim, Y.; Kwon, S.; Kim, M.; Kim, Y.H.; Kim, J.-H.; Park, T.J.; Kang, H.Y. Senescent fibroblasts drive ageing pigmentation: A potential therapeutic target for senile lentigo. Theranostics 2018, 8, 4620–4632. [Google Scholar] [CrossRef]
- Martic, I.; Wedel, S.; Jansen-Durr, P.; Cavinato, M. A new model to investigate UVB-induced cellular senescence and pigmentation in melanocytes. Mech. Ageing Dev. 2020, 190, 111322. [Google Scholar] [CrossRef] [PubMed]
- Bellei, B.; Picardo, M. Premature cell senescence in human skin: Dual face in chronic acquired pigmentary disorders. Ageing Res. Rev. 2020, 57, 100981. [Google Scholar] [CrossRef] [PubMed]
- Kovacs, D.; Bastonini, E.; Ottaviani, M.; Cota, C.; Migliano, E.; Dell’Anna, M.L.; Picardo, M. Vitiligo Skin: Exploring the Dermal Compartment. J. Investig. Dermatol. 2018, 138, 394–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patra, V.; Serezal, G.I.; Wolf, P. Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure. Nutrients 2020, 12, 1795. [Google Scholar] [CrossRef]
- Rong, J.; Shan, C.; Liu, S.; Zheng, H.; Liu, C.; Liu, M.; Jin, F.; Wang, L. Skin resistance to UVB-induced oxidative stress and hyperpigmentation by the topical use of Lactobacillus helveticus NS8-fermented milk supernatant. J. Appl. Microbiol. 2017, 123, 511–523. [Google Scholar] [CrossRef] [PubMed]
- Šahmatova, L.; Sügis, E.; Šunina, M.; Hermann, H.; Prans, E.; Pihlap, M.; Abram, K.; Rebane, A.; Peterson, H.; Peterson, P.; et al. Signs of innate immune activation and premature immunosenescence in psoriasis patients. Sci. Rep. 2017, 7, 7553. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.H.; Wu, C.S.; Chao, Y.H.; Lin, C.C.; Tsai, H.Y.; Li, Y.R.; Chen, Y.Z.; Tsai, W.H.; Chen, Y.K. Lactobacillus pentosus GMNL-77 inhibits skin lesions in imiquimod-induced psoriasis-like mice. J. Food Drug Anal. 2017, 25, 559–566. [Google Scholar] [CrossRef] [Green Version]
- Witte, M.; Thaçi, D. Psoriasis and the microbiome. Hautarzt 2019, 70, 416–421. [Google Scholar] [CrossRef]
- Tan, L.; Zhao, S.; Zhu, W.; Wu, L.; Li, J.; Shen, M.; Lei, L.; Chen, X.; Peng, C. The Akkermansia muciniphila is a gut microbiota signature in psoriasis. Exp. Dermatol. 2018, 27, 144–149. [Google Scholar] [CrossRef]
- Polak, K.; Bergler-Czop, B.; Szczepanek, M.; Wojciechowska, K.; Frątczak, A.; Kiss, N. Psoriasis and Gut Microbiome—Current State of Art. Int. J. Mol. Sci. 2021, 22, 4529. [Google Scholar] [CrossRef]
- Boothe, D.W.; Tarbox, J.A.; Tarbox, M.B. Atopic Dermatitis: Pathophysiology. In Management of Atopic Dermatitis: Methods and Challenges; Fortson, E.A., Feldman, S.R., Strowd, L.C., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 21–37. [Google Scholar]
- Dainichi, T.; Kitoh, A.; Otsuka, A.; Nakajima, S.; Nomura, T.; Kaplan, D.H.; Kabashima, K. The epithelial immune microenvironment (EIME) in atopic dermatitis and psoriasis. Nat. Immunol. 2018, 19, 1286–1298. [Google Scholar] [CrossRef]
- Krouwer, V.J.D.; Hekking, L.H.P.; Langelaar-Makkinje, M.; Regan-Klapisz, E.; Post, J.A. Endothelial cell senescence is associated with disrupted cell-cell junctions and increased monolayer permeability. Vasc. Cell 2012, 4, 12. [Google Scholar] [CrossRef] [Green Version]
- Polkowska-Pruszyńska, B.; Gerkowicz, A.; Krasowska, D. The gut microbiome alterations in allergic and inflammatory skin diseases—An update. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 455–464. [Google Scholar] [CrossRef]
- Thomas, C.L.; Fernández-Peñas, P. The microbiome and atopic eczema: More than skin deep. Australas. J. Dermatol. 2017, 58, 18–24. [Google Scholar] [CrossRef] [Green Version]
- McLaughlin, J.; Watterson, S.; Layton, A.M.; Bjourson, A.J.; Barnard, E.; McDowell, A. Propionibacterium acnes and Acne Vulgaris: New Insights from the Integration of Population Genetic, Multi-Omic, Biochemical and Host-Microbe Studies. Microorganisms 2019, 7, 128. [Google Scholar] [CrossRef] [Green Version]
- Tanghetti, E.A. The role of inflammation in the pathology of acne. J. Clin. Aesthet. Dermatol. 2013, 6, 27–35. [Google Scholar]
- Ribero, S.; Sanna, M.; Visconti, A.; Navarini, A.; Aviv, A.; Glass, D.; Spector, T.D.; Smith, C.; Simpson, M.; Barker, J.; et al. Acne and Telomere Length: A New Spectrum between Senescence and Apoptosis Pathways. J. Investig. Dermatol. 2017, 137, 513–515. [Google Scholar] [CrossRef] [Green Version]
- Askari, N.; Ghazanfari, T.; Yaraee, R.; Mahdavi, V.M.R.; Soroush, M.R.; Hassan, M.Z.; Khodashenas, Z.; Shams, J.; Faghihzadeh, S. Association between Acne and Serum Pro-inflammatory Cytokines (IL-1α, IL-1β, IL-1Ra, IL-6, IL-8, IL-12 and RANTES) in Mustard Gas-Exposed Patients: Sardasht-Iran Cohort Study. Arch. Iran. Med. 2017, 20, 86–91. [Google Scholar]
- Lee, Y.B.; Byun, E.J.; Kim, H.S. Potential Role of the Microbiome in Acne: A Comprehensive Review. J. Clin. Med. 2019, 8, 987. [Google Scholar] [CrossRef] [Green Version]
- Velnar, T.; Bailey, T.; Smrkolj, V. The wound healing process: An overview of the cellular and molecular mechanisms. J. Int. Med. Res. 2009, 37, 1528–1542. [Google Scholar] [CrossRef]
- Berg, V.J.S.; Rudolph, R.; Hollan, C.; Haywood-Reid, P.L. Fibroblast senescence in pressure ulcers. Wound Repair Regen. 1998, 6, 38–49. [Google Scholar] [CrossRef]
- Mendez, M.V.; Stanley, A.; Park, H.Y.; Shon, K.; Phillips, T.; Menzoian, J.O. Fibroblasts cultured from venous ulcers display cellular characteristics of senescence. J. Vasc. Surg. 1998, 28, 876–883. [Google Scholar] [CrossRef] [Green Version]
- Wilkinson, H.N.; Clowes, C.; Banyard, K.L.; Matteuci, P.; Mace, K.A.; Hardman, M.J. Elevated Local Senescence in Diabetic Wound Healing Is Linked to Pathological Repair via CXCR2. J. Investig. Dermatol. 2019, 139, 1171–1181.e6. [Google Scholar] [CrossRef]
- Wang, H.; Wang, Z.; Huang, Y.; Zhou, Y.; Sheng, X.; Jiang, Q.; Wang, Y.; Luo, P.; Luo, M.; Shi, C. Senolytics (DQ) Mitigates Radiation Ulcers by Removing Senescent Cells. Front. Oncol. 2019, 9, 1576. [Google Scholar] [CrossRef] [Green Version]
- Wilkinson, H.N.; Hardman, M.J. Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front. Cell Dev. Biol. 2020, 8, 773:1–773:13. [Google Scholar] [CrossRef]
- Drago, F.; Gariazzo, L.; Cioni, M.; Trave, I.; Parodi, A. The microbiome and its relevance in complex wounds. Eur. J. Dermatol. 2019, 29, 6–13. [Google Scholar] [CrossRef]
- Rahim, K.; Saleha, S.; Zhu, X.; Huo, L.; Basit, A.; Franco, O.L. Bacterial Contribution in Chronicity of Wounds. Microb. Ecol. 2017, 73, 710–721. [Google Scholar] [CrossRef]
- Attinger, C.; Wolcott, R. Clinically Addressing Biofilm in Chronic Wounds. Adv. Wound Care 2012, 1, 127–132. [Google Scholar] [CrossRef] [Green Version]
- Song, S.; Lam, E.W.; Tchkonia, T.; Kirkland, J.L.; Sun, Y. Senescent Cells: Emerging Targets for Human Aging and Age-Related Diseases. Trends Biochem. Sci. 2020, 45, 578–592. [Google Scholar] [CrossRef]
- Hadrup, S.R.; Strindhall, J.; Kollgaard, T.; Seremet, T.; Johansson, B.; Pawelec, G.; Straten, T.P.; Wikby, A. Longitudinal studies of clonally expanded CD8 T cells reveal a repertoire shrinkage predicting mortality and an increased number of dysfunctional cytomegalovirus-specific T cells in the very elderly. J. Immunol. 2006, 176, 2645–2653. [Google Scholar] [CrossRef] [Green Version]
- Ishihara, K.; Hirano, T. IL-6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine Growth Factor Rev. 2002, 13, 357–368. [Google Scholar] [CrossRef]
- Wu, H.-J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut. Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [Green Version]
- Amsterdam, D.; Ostrov, B.E. The Impact of the Microbiome on Immunosenescence. Immunol. Investig. 2018, 47, 801–811. [Google Scholar] [CrossRef] [PubMed]
- Atarashi, K.; Tanoue, T.; Ando, M.; Kamada, N.; Nagano, Y.; Narushima, S.; Suda, W.; Imaoka, A.; Setoyama, H.; Nagamori, T.; et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 2015, 163, 367–380. [Google Scholar] [CrossRef] [Green Version]
- Symons, A.; Budelsky, A.L.; Towne, J.E. Are Th17 cells in the gut pathogenic or protective? Mucosal Immunol. 2012, 5, 4–6. [Google Scholar] [CrossRef] [Green Version]
- Forever Healthy. Available online: https://brain.forever-healthy.org/display/EN/Dasatinib+and+Quercetin+Senolytic+Therapy (accessed on 10 November 2021).
- Kaefer, A.; Yang, J.; Noertersheuser, P.; Mensing, S.; Humerickhouse, R.; Awni, W.; Xiong, H. Mechanism-based pharmacokinetic/pharmacodynamic meta-analysis of navitoclax (ABT-263) induced thrombocytopenia. Cancer Chemother. Pharmacol. 2014, 74, 593–602. [Google Scholar] [CrossRef]
- He, Y.; Zhang, X.; Chang, J.; Kim, H.-N.; Zhang, P.; Wang, Y.; Khan, S.; Liu, X.; Zhang, X.; Lv, D.; et al. Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat. Commun. 2020, 11, 1996. [Google Scholar] [CrossRef]
- Leverson, J.D.; Phillips, D.C.; Mitten, M.J.; Boghaert, E.R.; Diaz, D.; Tahir, S.K.; Belmont, L.D.; Nimmer, P.; Xiao, Y.; Ma, X.M.; et al. Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy. Sci. Transl. Med. 2015, 7, 279ra240. [Google Scholar] [CrossRef] [PubMed]
- Blagosklonny, M.V. Rapamycin for longevity: Opinion article. Aging 2019, 11, 8048–8067. [Google Scholar] [CrossRef]
- Espada, L.; Dakhovnik, A.; Chaudhari, P.; Martirosyan, A.; Miek, L.; Poliezhaieva, T.; Schaub, Y.; Nair, A.; Döring, N.; Rahnis, N.; et al. Loss of metabolic plasticity underlies metformin toxicity in aged Caenorhabditis elegans. Nature 2020, 2, 1316–1331. [Google Scholar] [CrossRef]
- Glossmann, H.H.; Lutz, O.M.D. Metformin and Aging: A Review. Gerontology 2019, 65, 581–590. [Google Scholar] [CrossRef] [PubMed]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [Green Version]
- Hickson, L.J.; Prata, L.L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [Green Version]
- Yosef, R.; Pilpel, N.; Tokarsky-Amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [Google Scholar] [CrossRef] [PubMed]
- Borras, C.; Abdelaziz, K.M.; Gambini, J.; Serna, E.; Inglés, M.; de la Fuente, M.; Garcia, I.; Matheu, A.; Sanchís, P.; Belenguer, A.; et al. Human exceptional longevity: Transcriptome from centenarians is distinct from septuagenarians and reveals a role of Bcl-xL in successful aging. Aging 2016, 8, 3185–3208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feins, S.; Kong, W.; Williams, E.F.; Milone, M.C.; Fraietta, J.A. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am. J. Hematol. 2019, 94, S3–S9. [Google Scholar] [CrossRef] [Green Version]
- Amor, C.; Feucht, J.; Leibold, J.; Ho, Y.-J.; Zhu, C.; Alonso-Curbelo, D.; Mansilla-Soto, J.; Boyer, J.A.; Li, X.; Giavridis, T.; et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020, 583, 127–132. [Google Scholar] [CrossRef]
- Betz, C.; Hall, M.N. Where is mTOR and what is it doing there? J. Cell Biol. 2013, 203, 563–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laberge, R.M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef] [PubMed]
- Weichhart, T. mTOR as Regulator of Lifespan, Aging, and Cellular Senescence: A Mini-Review. Gerontology 2018, 64, 127–134. [Google Scholar] [CrossRef]
- Bitto, A.; Ito, T.K.; Pineda, V.V.; LeTexier, N.J.; Huang, H.Z.; Sutlief, E.; Tung, H.; Vizzini, N.; Chen, B.; Smith, K.; et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife 2016, 5, e16351. [Google Scholar] [CrossRef]
- Harrison, D.E.; Strong, R.; Sharp, Z.D.; Nelson, J.F.; Astle, C.M.; Flurkey, K.; Nadon, N.L.; Wilkinson, J.E.; Frenkel, K.; Carter, C.S.; et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 2009, 460, 392–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moiseeva, O.; Deschênes-Simard, X.; St-Germain, E.; Igelmann, S.; Huot, G.; Cadar, A.E.; Bourdeau, V.; Pollak, M.N.; Ferbeyre, G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef] [PubMed]
- Śmieszek, A.; Stręk, Z.; Kornicka, K.; Grzesiak, J.; Weiss, C.; Marycz, K. Antioxidant and Anti-Senescence Effect of Metformin on Mouse Olfactory Ensheathing Cells (mOECs) May Be Associated with Increased Brain-Derived Neurotrophic Factor Levels-An Ex Vivo Study. Int. J. Mol. Sci. 2017, 18, 872. [Google Scholar] [CrossRef]
- Espinoza, S.E.; Musi, N.; Wang, C.P.; Michalek, J.; Orsak, B.; Romo, T.; Powers, B.; Conde, A.; Moris, M.; Bair-Kelps, D.; et al. Rationale and Study Design of a Randomized Clinical Trial of Metformin to Prevent Frailty in Older Adults with Prediabetes. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 102–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Correia-Melo, C.; Marques, F.D.; Anderson, R.; Hewitt, G.; Hewitt, R.; Cole, J.; Carroll, B.M.; Miwa, S.; Birch, J.; Merz, A.; et al. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 2016, 35, 724–742. [Google Scholar] [CrossRef]
- Kaur, A.; Macip, S.; Stover, C.M. An Appraisal on the Value of Using Nutraceutical Based Senolytics and Senostatics in Aging. Front. Cell Dev. Biol. 2020, 8, 218:1–218:9. [Google Scholar] [CrossRef]
- David, A.A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef] [Green Version]
- Soto-Gamez, A.; Demaria, M. Therapeutic interventions for aging: The case of cellular senescence. Drug Discov. 2017, 22, 786–795. [Google Scholar] [CrossRef]
- An, S.; Cho, S.-Y.; Kang, J.; Lee, S.; Kim, H.-S.; Min, D.-J.; Son, E.; Cho, K.-H. Inhibition of 3-phosphoinositide–dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts. Proc. Natl. Acad. Sci. USA 2020, 117, 31535–31546. [Google Scholar] [CrossRef]
- Fyffe, C.; Falasca, M. 3-Phosphoinositide-dependent protein kinase-1 as an emerging target in the management of breast cancer. Cancer Manag. Res. 2013, 5, 271–280. [Google Scholar] [CrossRef] [Green Version]
- Chondrogianni, N.; Kapeta, S.; Chinou, I.; Vassilatou, K.; Papassideri, I.; Gonos, E.S. Anti-ageing and rejuvenating effects of quercetin. Exp. Gerontol. 2010, 45, 763–771. [Google Scholar] [CrossRef] [Green Version]
- Lewinska, A.; Adamczyk-Grochala, J.; Bloniarz, D.; Olszowka, J.; Kulpa-Greszta, M.; Litwinienko, G.; Tomaszewska, A.; Wnuk, M.; Pazik, R. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe3O4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox. Biol. 2020, 28, 101337. [Google Scholar] [CrossRef] [PubMed]
- Moon, K.-C.; Yang, J.-P.; Lee, J.-S.; Jeong, S.-H.; Dhong, E.-S.; Han, S.-K. Effects of Ultraviolet Irradiation on Cellular Senescence in Keratinocytes Versus Fibroblasts. J. Craniofac. Surg. 2019, 30, 270–275. [Google Scholar] [CrossRef]
- Shin, E.J.; Lee, J.S.; Hong, S.; Lim, T.-G.; Byun, S. Quercetin Directly Targets JAK2 and PKCδ and Prevents UV-Induced Photoaging in Human Skin. Int. J. Mol. Sci. 2019, 20, 5262. [Google Scholar] [CrossRef] [Green Version]
- Khan, N.; Syed, D.N.; Ahmad, N.; Mukhtar, H. Fisetin: A Dietary Antioxidant for Health Promotion. Antioxid. Redox. Signal 2012, 19, 151–162. [Google Scholar] [CrossRef] [PubMed]
- Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsoukalas, D.; Fragkiadaki, P.; Docea, A.O.; Alegakis, A.K.; Sarandi, E.; Vakonaki, E.; Salataj, E.; Kouvidi, E.; Nikitovic, D.; Kovatsi, L.; et al. Association of nutraceutical supplements with longer telomere length. Int. J. Mol. Med. 2019, 44, 218–226. [Google Scholar] [CrossRef] [Green Version]
- Mack, D.R. Probiotics-mixed messages. Can. Fam. Physician 2005, 51, 1455–1464. [Google Scholar]
- Sharma, R.; Padwad, Y. Probiotic bacteria as modulators of cellular senescence: Emerging concepts and opportunities. Gut Microbes 2020, 11, 335–349. [Google Scholar] [CrossRef]
- Westfall, S.; Lomis, N.; Prakash, S. Longevity extension in Drosophila through gut-brain communication. Sci. Rep. 2018, 8, 8362. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; Sharma, A.; Gupta, M.; Padwad, Y.; Sharma, R. Cell-Free Culture Supernatant of Probiotic Lactobacillus fermentum Protects Against H2O2-Induced Premature Senescence by Suppressing ROS-Akt-mTOR Axis in Murine Preadipocytes. Probiotics Antimicrob. 2020, 12, 563–576. [Google Scholar] [CrossRef] [PubMed]
- Quan, T.; Fisher, G.J. Role of Age-Associated Alterations of the Dermal Extracellular Matrix Microenvironment in Human Skin Aging: A Mini-Review. Gerontology 2015, 61, 427–434. [Google Scholar] [CrossRef] [Green Version]
- Kohl, E.; Steinbauer, J.; Landthaler, M.; Szeimies, R.-M. Skin ageing. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 873–884. [Google Scholar] [CrossRef] [PubMed]
- Im, A.R.; Lee, B.; Kang, D.J.; Chae, S. Protective effects of tyndallized Lactobacillus acidophilus IDCC 3302 against UVBinduced photodamage to epidermal keratinocytes cells. Int. J. Mol. Med. 2019, 43, 2499–2506. [Google Scholar] [CrossRef]
- Lim, H.Y.; Jeong, D.; Park, S.H.; Shin, K.K.; Hong, Y.H.; Kim, E.; Yu, Y.-G.; Kim, T.-R.; Kim, H.; Lee, J.; et al. Antiwrinkle and Antimelanogenesis Effects of Tyndallized Lactobacillus acidophilus KCCM12625P. Int. J. Mol. Sci. 2020, 21, 1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christensen, G.J.; Brüggemann, H. Bacterial skin commensals and their role as host guardians. Benef. Microbes 2014, 5, 201–215. [Google Scholar] [CrossRef] [PubMed]
- Kholmanskikh, O.; van Baren, N.; Brasseur, F.; Ottaviani, S.; Vanacker, J.; Arts, N.; van der Bruggen, P.; Coulie, P.; De Plaen, E. Interleukins 1alpha and 1beta secreted by some melanoma cell lines strongly reduce expression of MITF-M and melanocyte differentiation antigens. Int. J. Cancer 2010, 127, 1625–1636. [Google Scholar] [CrossRef]
- Chen, N.; Hu, Y.; Li, W.H.; Eisinger, M.; Seiberg, M.; Lin, C.B. The role of keratinocyte growth factor in melanogenesis: A possible mechanism for the initiation of solar lentigines. Exp. Dermatol. 2010, 19, 865–872. [Google Scholar] [CrossRef] [PubMed]
- Moretti, S.; Spallanzani, A.; Amato, L.; Hautmann, G.; Gallerani, I.; Fabiani, M.; Fabbri, P. New insights into the pathogenesis of vitiligo: Imbalance of epidermal cytokines at sites of lesions. Pigment. Cell Res. 2002, 15, 87–92. [Google Scholar] [CrossRef]
- Goodarzi, A.; Mozafarpoor, S.; Bodaghabadi, M.; Mohamadi, M. The potential of probiotics for treating acne vulgaris: A review of literature on acne and microbiota. Dermatol. Ther. 2020, 33, e13279. [Google Scholar] [CrossRef] [PubMed]
- Bowe, W.P.; Filip, J.C.; DiRienzo, J.M.; Volgina, A.; Margolis, D.J. Inhibition of propionibacterium acnes by bacteriocin-like inhibitory substances (BLIS) produced by Streptococcus salivarius. J. Drugs Dermatol. 2006, 5, 868–870. [Google Scholar] [PubMed]
- Ridiandries, A.; Tan, J.T.M.; Bursill, C.A. The Role of Chemokines in Wound Healing. Int. J. Mol. Sci. 2018, 19, 3217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jun, J.-I.; Lau, L.F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef] [PubMed]
- Lindley, L.E.; Stojadinovic, O.; Pastar, I.; Tomic-Canic, M. Biology and Biomarkers for Wound Healing. Plast. Reconstr. Surg. 2016, 138, 18S–28S. [Google Scholar] [CrossRef] [PubMed]
- Lai, Y.; Di Nardo, A.; Nakatsuji, T.; Leichtle, A.; Yang, Y.; Cogen, A.L.; Wu, Z.R.; Hooper, L.V.; Schmidt, R.R.; von Aulock, S.; et al. Commensal bacteria regulate Toll-like receptor 3-dependent inflammation after skin injury. Nat. Med. 2009, 15, 1377–1382. [Google Scholar] [CrossRef]
- Naik, S.; Bouladoux, N.; Linehan, J.L.; Han, S.-J.; Harrison, O.J.; Wilhelm, C.; Conlan, S.; Himmelfarb, S.; Byrd, A.L.; Deming, C.; et al. Commensal–dendritic-cell interaction specifies a unique protective skin immune signature. Nature 2015, 520, 104–108. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, S. Emerging Insights into the Metabolic Alterations in Aging Using Metabolomics. Metabolites 2019, 9, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chung, H.-J.; Sim, J.-H.; Min, T.-S.; Choi, H.-K. Metabolomics and Lipidomics Approaches in the Science of Probiotics: A Review. J. Med. Food 2018, 21, 1086–1095. [Google Scholar] [CrossRef] [PubMed]
- Avanesov, A.S.; Ma, S.; Pierce, K.A.; Yim, S.H.; Lee, B.C.; Clish, C.B.; Gladyshev, V.N. Age- and diet-associated metabolome remodeling characterizes the aging process driven by damage accumulation. Elife 2014, 3, e02077. [Google Scholar] [CrossRef]
- Sharma, R.; Ramanathan, A. The Aging Metabolome—Biomarkers to Hub Metabolites. Proteomics 2020, 20, 1800407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nacarelli, T.; Lau, L.; Fukumoto, T.; Zundell, J.; Fatkhutdinov, N.; Wu, S.; Aird, K.M.; Iwasaki, O.; Kossenkov, A.V.; Schultz, D.; et al. NAD(+) metabolism governs the proinflammatory senescence-associated secretome. Nat. Cell Biol. 2019, 21, 397–407. [Google Scholar] [CrossRef] [PubMed]
- Elpa, D.P.; Chiu, H.-Y.; Wu, S.-P.; Urban, P.L. Skin Metabolomics. Trends Endocrinol. Metab. 2021, 32, 66–75. [Google Scholar] [CrossRef] [PubMed]
- Brasili, E.; Mengheri, E.; Tomassini, A.; Capuani, G.; Roselli, M.; Finamore, A.; Sciubba, F.; Marini, F.; Miccheli, A. Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12 Induce Different Age-Related Metabolic Profiles Revealed by 1H-NMR Spectroscopy in Urine and Feces of Mice. J. Nutr. 2013, 143, 1549–1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- BCC Research. Available online: https://www.bccresearch.com/market-research/biotechnology/global-longevity-and-anti-senescence-therapy-market-report.html (accessed on 10 November 2021).
- Research and Markets. Available online: https://www.globenewswire.com/news-release/2020/07/24/2067180/0/en/Anti-Aging-Products-Industry-Projected-to-be-Worth-83-2-Billion-by-2027-Key-Trends-Opportunities-and-Players.html (accessed on 10 November 2021).
- Research and Markets. Available online: https://www.researchandmarkets.com/reports/4766817/global-probiotic-supplements-market-2018-2027?utm_source=BW&utm_medium=PressRelease&utm_code=2ldqfg&utm_campaign=1239349+-+Global+%243.28+Billion+Probiotic+Supplements+Market+(2018-2027)%3a+Development+of+Dry+Probiotic+Supplements+for+Chronic+Diseases+will+Fuel+Growth+at+an+Expected+CAGR+of+10.15%25+During+2018-2027&utm_exec=joca220prd (accessed on 10 November 2021).
- Fortune Business Insights. Available online: https://www.globenewswire.com/news-release/2021/02/17/2176980/0/en/Skincare-Market-Revenue-to-Hit-USD-200-25-Billion-by-2026-Driven-by-the-Increasing-Investment-in-Product-R-D-says-Fortune-Business-Insights.html (accessed on 10 November 2021).
Pharmaceutical Agent | Mechanism | Long-Term Side Effects |
---|---|---|
Dasatinib + Quercetin | Senolytic | Currently unknown with further investigation required [129]. |
Navitoclax | Senolytic | Platelet toxicity or thrombocytopenia [130,131]; neutrophil toxicity [132]. |
Rapamycin | Senostatic | Glucose intolerance; insulin resistance; hyperglycemia; starvation pseudo-diabetes; stomatitis; mucositis; interstitial pneumonitis [133]. |
Metformin | Senostatic | Shortened lifespan; mitochondrial dysfunction; lethal ATP exhaustion [134,135]. |
Skin Disease | SASP Factor(s) Involved | Probiotic Influence on Factor(s) and Disease |
---|---|---|
Aging | Upregulated: |
|
Carcinogenesis | Upregulated: |
|
Dyspigmentation | Modulated: |
|
Psoriasis and Atopic Dermatitis | Upregulated: |
|
Acne Vulgaris | Upregulated: | |
Chronic Wounds | Upregulated: |
|
Immunity Decline | Dysregulated: |
|
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Boyajian, J.L.; Ghebretatios, M.; Schaly, S.; Islam, P.; Prakash, S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients 2021, 13, 4550. https://doi.org/10.3390/nu13124550
Boyajian JL, Ghebretatios M, Schaly S, Islam P, Prakash S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients. 2021; 13(12):4550. https://doi.org/10.3390/nu13124550
Chicago/Turabian StyleBoyajian, Jacqueline Lena, Merry Ghebretatios, Sabrina Schaly, Paromita Islam, and Satya Prakash. 2021. "Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence" Nutrients 13, no. 12: 4550. https://doi.org/10.3390/nu13124550
APA StyleBoyajian, J. L., Ghebretatios, M., Schaly, S., Islam, P., & Prakash, S. (2021). Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients, 13(12), 4550. https://doi.org/10.3390/nu13124550