Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models
Abstract
:1. Introduction
2. The Transcription Factor NF-κB
3. Role of NF-κB in Ageing
3.1. Pathways Related to Ageing That Converge in the Regulation of NF-κB Activation
3.2. Role of NF-κB in the Maintenance and Reversion of the Ageing State
4. Study of Age-Related Diseases through the Analysis of Genetically Modified Mouse Models of the NF-κB Signaling Pathways
4.1. A Tight Relationship between NF-κB Activation-Inflammation- and Age-Associated Diseases
4.2. Involvement of NF-κB in Age-Associated Eye Diseases
4.3. Role of NF-κB in Age-Related Neurological Diseases
4.4. Senescent Osteoarthritis and Osteoporosis
4.5. Role of NF-κB in Age-Associated Muscle Diseases: Muscle Wasting, Sarcopenia, Cachexia
4.6. Implication of NF-κB in Age-Related Heart Diseases
4.6.1. Heart Failure, and Cardiac Hypertrophy and Remodeling
4.6.2. Atherosclerosis
4.7. Role of NF-κB Activation in the Development of Age-Associated Diseases with High Risk of Cancer Development
4.7.1. NF-κB in Cardiometabolic Disorders: Obesity, Insulin Resistance, Type II Diabetes Mellitus
4.7.2. Age-Related Liver Diseases
5. Role of NF-κB in the Decline of Stem Cell Functions in Aged Individuals
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
Abbreviations
References
- Kenyon, C.J. The genetics of ageing. Nature 2010, 464, 504–512. [Google Scholar] [CrossRef]
- Jin, K.; Simpkins, J.W.; Ji, X.; Leis, M.; Stambler, I. The Critical Need to Promote Research of Aging and Aging-related Diseases to Improve Health and Longevity of the Elderly Population. Aging Dis. 2015, 6, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tilstra, J.S.; Clauson, C.L.; Niedernhofer, L.J.; Robbins, P.D. NF-kB in Aging and Disease. Aging Dis. 2011, 2, 449–465. [Google Scholar] [PubMed]
- Hayden, M.S.; West, A.P.; Ghosh, S. NF-kappaB and the immune response. Oncogene 2006, 25, 6758–6780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munn, L.L. Cancer and inflammation. Wiley interdisciplinary reviews. Syst. Biol. Med. 2017, 9, e1370. [Google Scholar] [CrossRef] [Green Version]
- Salminen, A.; Kaarniranta, K. NF-kappaB signaling in the aging process. J. Clin. Immunol. 2009, 29, 397–405. [Google Scholar] [CrossRef] [PubMed]
- Osorio, F.G.; Soria-Valles, C.; Santiago-Fernández, O.; Freije, J.M.P.; López-Otín, C. NF-kappaB signaling as a driver of ageing. Int. Rev. Cell Mol. Biol. 2016, 326, 133–174. [Google Scholar] [CrossRef]
- Folgueras, A.R.; Freitas-Rodríguez, S.; Velasco, G.; López-Otín, C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ. Res. 2018, 123, 905–924. [Google Scholar] [CrossRef]
- Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
- Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [Green Version]
- Ghosh, S.; Karin, M. Missing pieces in the NF-kappaB puzzle. Cell 2002, 109, S81–S96. [Google Scholar] [CrossRef] [Green Version]
- Silverman, N.; Maniatis, T. NF-kappaB signaling pathways in mammalian and insect innate immunity. Genes Dev. 2001, 15, 2321–2342. [Google Scholar] [CrossRef] [Green Version]
- Pérez, P.; Page, A.; Jorcano, J.L. Role of phosphorylated p50-NF-kappaB in the ultraviolet response of mouse skin. Mol. Carcinog. 2000, 27, 272–279. [Google Scholar] [CrossRef]
- Massoumi, R.; Chmielarska, K.; Hennecke, K.; Pfeifer, A.; Fässler, R. Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent NF-kappaB signaling. Cell 2006, 125, 665–677. [Google Scholar] [CrossRef] [Green Version]
- Kerr, L.D.; Duckett, C.S.; Wamsley, P.; Zhang, Q.; Chiao, P.; Nabel, G.; McKeithan, T.W.; Baeuerle, P.A.; Verma, I.M. The proto-oncogene bcl-3 encodes an I kappa B protein. Genes Dev. 1992, 6, 2352–2363. [Google Scholar] [CrossRef] [Green Version]
- Hayden, M.S.; Ghosh, S. NF-κB, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 2012, 26, 203–234. [Google Scholar] [CrossRef] [Green Version]
- Hayden, M.S.; Ghosh, S. Signaling to NF-kappaB. Genes Dev. 2004, 18, 2195–2224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Q.; Verma, I.M. NF-kappaB regulation in the immune system. Nat. Rev. Immunol. 2002, 2, 725–734. [Google Scholar] [CrossRef] [PubMed]
- Delhase, M.; Karin, M. The I kappa B kinase: A master regulator of NF-kappa B, innate immunity, and epidermal differentiation. Cold Spring Harb. Symp. Quant. Biol. 1999, 64, 491–503. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, M.; Fuentes, M.E.; Yamaguchi, K.; Durnin, M.H.; Dalrymple, S.A.; Hardy, K.L.; Goeddel, D.V. Embryonic lethality, liver degeneration, and impaired NF-kappa B activation in IKK-beta-deficient mice. Immunity 1999, 10, 421–429. [Google Scholar] [CrossRef] [Green Version]
- Rudolph, D.; Yeh, W.C.; Wakeham, A.; Rudolph, B.; Nallainathan, D.; Potter, J.; Elia, A.J.; Mak, T.W. Severe liver degeneration and lack of NF-kappaB activation in NEMO/IKKgamma-deficient mice. Genes Dev. 2000, 14, 854–862. [Google Scholar]
- Israël, A. The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harb. Perspect. Biol. 2010, 2, a000158. [Google Scholar] [CrossRef] [Green Version]
- Huang, W.C.; Hung, M.C. Beyond NF-κB activation: Nuclear functions of IκB kinase α. J. Biomed. Sci. 2013, 20, 3. [Google Scholar] [CrossRef] [Green Version]
- Régnier, C.H.; Song, H.Y.; Gao, X.; Goeddel, D.V.; Cao, Z.; Rothe, M. Identification and characterization of an IkappaB kinase. Cell 1997, 90, 373–383. [Google Scholar] [CrossRef] [Green Version]
- Bonizzi, G.; Karin, M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004, 25, 280–288. [Google Scholar] [CrossRef]
- Yamamoto, Y.; Gaynor, R.B. IkappaB kinases: Key regulators of the NF-kappaB pathway. Trends Biochem. Sci. 2004, 29, 72–79. [Google Scholar] [CrossRef] [PubMed]
- Senftleben, U.; Cao, Y.; Xiao, G.; Greten, F.R.; Krahn, G.; Bonizzi, G.; Chen, Y.; Hu, Y.; Fong, A.; Sun, S.C.; et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 2001, 293, 1495–1499. [Google Scholar] [CrossRef] [PubMed]
- Cao, Y.; Bonizzi, G.; Seagroves, T.N.; Greten, F.R.; Johnson, R.; Schmidt, E.V.; Karin, M. IKKalpha provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell 2001, 107, 763–775. [Google Scholar] [CrossRef] [Green Version]
- Song, H.Y.; Rothe, M.; Goeddel, D.V. The tumor necrosis factor-inducible zinc finger protein A20 interacts with TRAF1/TRAF2 and inhibits NF-kappaB activation. Proc. Natl. Acad. Sci. USA 1996, 93, 6721–6725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brummelkamp, T.R.; Nijman, S.M.; Dirac, A.M.; Bernards, R. Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappaB. Nature 2003, 424, 797–801. [Google Scholar] [CrossRef]
- Kovalenko, A.; Chable-Bessia, C.; Cantarella, G.; Israel, A.; Wallach, D.; Courtois, G. The tumour suppressor CYLD negatively regulates NF-kappaB signalling by deubiquitination. Nature 2003, 424, 801–805. [Google Scholar] [CrossRef]
- Trompouki, E.; Hatzivassiliou, E.; Tsichritzis, T.; Farmer, H.; Ashworth, A.; Mosialos, G. CYLD is a deubiquitinating enzyme that negatively regulates NF-kappaB activation by TNFR family members. Nature 2003, 424, 793–796. [Google Scholar] [CrossRef]
- Schuster, M.; Annemann, M.; Plaza-Sirvent, C.; Schmitz, I. Atypical IκB proteins-nuclear modulators of NF-κB signaling. Cell Commun. Signal CCS 2013, 11, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madrid, L.V.; Mayo, M.W.; Reuther, J.Y.; Baldwin, A.S., Jr. Akt stimulates the transactivation potential of the RelA/p65 Subunit of NF-kappa B through utilization of the Ikappa B kinase and activation of the mitogen-activated protein kinase p38. J. Biol. Chem. 2001, 276, 18934–18940. [Google Scholar] [CrossRef] [Green Version]
- Madrid, L.V.; Wang, C.Y.; Guttridge, D.C.; Schottelius, A.J.; Baldwin, A.S., Jr.; Mayo, M.W. Akt suppresses apoptosis by stimulating the transactivation potential of the RelA/p65 subunit of NF-kappaB. Mol. Cell Biol. 2000, 20, 1626–1638. [Google Scholar] [CrossRef] [Green Version]
- Petroulakis, E.; Mamane, Y.; Le Bacquer, O.; Shahbazian, D.; Sonenberg, N. mTOR signaling: Implications for cancer and anticancer therapy. Br. J. Cancer 2006, 94, 195–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vellai, T.; Takacs-Vellai, K.; Zhang, Y.; Kovacs, A.L.; Orosz, L.; Müller, F. Genetics: Influence of TOR kinase on lifespan in C. elegans. Nature 2003, 426, 620. [Google Scholar] [CrossRef]
- Dan, H.C.; Cooper, M.J.; Cogswell, P.C.; Duncan, J.A.; Ting, J.P.; Baldwin, A.S. Akt-dependent regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with IKK. Genes Dev. 2008, 22, 1490–1500. [Google Scholar] [CrossRef] [Green Version]
- Veuger, S.J.; Durkacz, B.W. Persistence of unrepaired DNA double strand breaks caused by inhibition of ATM does not lead to radio-sensitisation in the absence of NF-κB activation. DNA Repair 2011, 10, 235–244. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.H.; Shi, Y.; Tibbetts, R.S.; Miyamoto, S. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science 2006, 311, 1141–1146. [Google Scholar] [CrossRef]
- Choudhury, A.R.; Ju, Z.; Djojosubroto, M.W.; Schienke, A.; Lechel, A.; Schaetzlein, S.; Jiang, H.; Stepczynska, A.; Wang, C.; Buer, J.; et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat. Genet. 2007, 39, 99–105. [Google Scholar] [CrossRef]
- Lee, W.P.; Hou, M.C.; Lan, K.H.; Li, C.P.; Chao, Y.; Lin, H.C.; Lee, S.D. Helicobacter pylori-induced chronic inflammation causes telomere shortening of gastric mucosa by promoting PARP-1-mediated non-homologous end joining of DNA. Arch. Biochem. Biophys. 2016, 606, 90–98. [Google Scholar] [CrossRef]
- Wu, X.Q.; Yang, Y.; Li, W.X.; Cheng, Y.H.; Li, X.F.; Huang, C.; Meng, X.M.; Wu, B.M.; Liu, X.H.; Zhang, L.; et al. Telomerase reverse transcriptase acts in a feedback loop with NF-κB pathway to regulate macrophage polarization in alcoholic liver disease. Sci. Rep. 2016, 6, 18685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grabowska, W.; Sikora, E.; Bielak-Zmijewska, A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology 2017, 18, 447–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [Green Version]
- Kawahara, T.L.; Michishita, E.; Adler, A.S.; Damian, M.; Berber, E.; Lin, M.; McCord, R.A.; Ongaigui, K.C.; Boxer, L.D.; Chang, H.Y.; et al. SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal life span. Cell 2009, 136, 62–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.J.; Jung, K.J.; Yu, B.P.; Cho, C.G.; Choi, J.S.; Chung, H.Y. Modulation of redox-sensitive transcription factors by calorie restriction during aging. Mech. Ageing Dev. 2002, 123, 1589–1595. [Google Scholar] [CrossRef]
- Adler, A.S.; Sinha, S.; Kawahara, T.L.; Zhang, J.Y.; Segal, E.; Chang, H.Y. Motif module map reveals enforcement of aging by continual NF-kappaB activity. Genes Dev. 2007, 21, 3244–3257. [Google Scholar] [CrossRef] [Green Version]
- Kriete, A.; Mayo, K.L.; Yalamanchili, N.; Beggs, W.; Bender, P.; Kari, C.; Rodeck, U. Cell autonomous expression of inflammatory genes in biologically aged fibroblasts associated with elevated NF-kappaB activity. Immun. Ageing I A 2008, 5, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brégégère, F.; Milner, Y.; Friguet, B. The ubiquitin-proteasome system at the crossroads of stress-response and ageing pathways: A handle for skin care? Ageing Res. Rev. 2006, 5, 60–90. [Google Scholar] [CrossRef]
- Giardina, C.; Hubbard, A.K. Growing old with nuclear factor-kappaB. Cell Stress Chaperones 2002, 7, 207–212. [Google Scholar] [CrossRef]
- Helenius, M.; Hänninen, M.; Lehtinen, S.K.; Salminen, A. Changes associated with aging and replicative senescence in the regulation of transcription factor nuclear factor-kappa B. Biochem. J. 1996, 318, 603–608. [Google Scholar] [CrossRef]
- Cai, D.; Frantz, J.D.; Tawa, N.E., Jr.; Melendez, P.A.; Oh, B.C.; Lidov, H.G.; Hasselgren, P.O.; Frontera, W.R.; Lee, J.; Glass, D.J.; et al. IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. Cell 2004, 119, 285–298. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Drozdov, I.; Shroff, R.; Beltran, L.E.; Shanahan, C.M. Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells. Circ. Res. 2013, 112, e99–e109. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Liu, K.; Robinson, A.R.; Clauson, C.L.; Blair, H.C.; Robbins, P.D.; Niedernhofer, L.J.; Ouyang, H. DNA damage drives accelerated bone aging via an NF-kappaB-dependent mechanism. J. Bone Miner. Res. 2013, 28, 1214–1228. [Google Scholar] [CrossRef]
- Valen, G. Signal transduction through nuclear factor kappa B in ischemia-reperfusion and heart failure. Basic Res. Cardiol. 2004, 99, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Castorina, A.; Szychlinska, M.A.; Marzagalli, R.; Musumeci, G. Mesenchymal stem cells-based therapy as a potential treatment in neurodegenerative disorders: Is the escape from senescence an answer? Neural Regen. Res. 2015, 10, 850–858. [Google Scholar] [CrossRef] [PubMed]
- Meyerovich, K.; Ortis, F.; Allagnat, F.; Cardozo, A.K. Endoplasmic reticulum stress and the unfolded protein response in pancreatic islet inflammation. J. Mol. Endocrinol. 2016, 57, R1–R17. [Google Scholar] [CrossRef] [Green Version]
- Berenbaum, F. Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthr. Cartil. 2013, 21, 16–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steinman, L. Nuanced roles of cytokines in three major human brain disorders. J. Clin. Investig. 2008, 118, 3557–3563. [Google Scholar] [CrossRef]
- Ghosh, A.; Roy, A.; Liu, X.; Kordower, J.H.; Mufson, E.J.; Hartley, D.M.; Ghosh, S.; Mosley, R.L.; Gendelman, H.E.; Pahan, K. Selective inhibition of NF-kappaB activation prevents dopaminergic neuronal loss in a mouse model of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2007, 104, 18754–18759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ju Hwang, C.; Choi, D.Y.; Park, M.H.; Hong, J.T. NF-κB as a Key Mediator of Brain Inflammation in Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2019, 18, 3–10. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.S.; Rai, S.N.; Birla, H.; Zahra, W.; Rathore, A.S.; Singh, S.P. NF-κB-Mediated Neuroinflammation in Parkinson’s Disease and Potential Therapeutic Effect of Polyphenols. Neurotox. Res. 2020, 37, 491–507. [Google Scholar] [CrossRef] [PubMed]
- Bernal, G.M.; Wahlstrom, J.S.; Crawley, C.D.; Cahill, K.E.; Pytel, P.; Liang, H.; Kang, S.; Weichselbaum, R.R.; Yamini, B. Loss of Nfkb1 leads to early onset aging. Aging 2014, 6, 931–943. [Google Scholar] [CrossRef] [Green Version]
- Jurk, D.; Wilson, C.; Passos, J.F.; Oakley, F.; Correia-Melo, C.; Greaves, L.; Saretzki, G.; Fox, C.; Lawless, C.; Anderson, R.; et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2014, 2, 4172. [Google Scholar] [CrossRef] [PubMed]
- Alameda, J.P.; Ramirez, A.; Garcia-Fernandez, R.A.; Navarro, M.; Page, A.; Segovia, J.C.; Sanchez, R.; Suarez-Cabrera, C.; Paramio, J.M.; Bravo, A.; et al. Premature aging and cancer development in transgenic mice lacking functional CYLD. Aging 2019, 11, 127–159. [Google Scholar] [CrossRef]
- Page, A.; Cascallana, J.L.; Casanova, M.L.; Navarro, M.; Alameda, J.P.; Pérez, P.; Bravo, A.; Ramírez, A. IKKβ overexpression leads to pathologic lesions in stratified epithelia and exocrine glands and to tumoral transformation of oral epithelia. Mol. Cancer Res. 2011, 9, 1329–1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Osorio, F.G.; Barcena, C.; Soria-Valles, C.; Ramsay, A.J.; de Carlos, F.; Cobo, J.; Fueyo, A.; Freije, J.M.; Lopez-Otin, C. Nuclear lamina defects cause ATM-dependent NF-kappaB activation and link accelerated aging to a systemic inflammatory response. Genes Dev. 2012, 26, 2311–2324. [Google Scholar] [CrossRef] [Green Version]
- Moskalev, A.; Shaposhnikov, M. Pharmacological inhibition of NF-κB prolongs lifespan of Drosophila melanogaster. Aging 2011, 3, 391–394. [Google Scholar] [CrossRef]
- Strong, R.; Miller, R.A.; Astle, C.M.; Floyd, R.A.; Flurkey, K.; Hensley, K.L.; Javors, M.A.; Leeuwenburgh, C.; Nelson, J.F.; Ongini, E.; et al. Nordihydroguaiaretic acid and aspirin increase lifespan of genetically heterogeneous male mice. Aging Cell 2008, 7, 641–650. [Google Scholar] [CrossRef] [Green Version]
- Mostoslavsky, R.; Chua, K.F.; Lombard, D.B.; Pang, W.W.; Fischer, M.R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M.M.; et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 2006, 124, 315–329. [Google Scholar] [CrossRef] [Green Version]
- Soria-Valles, C.; Osorio, F.G.; Gutiérrez-Fernández, A.; De Los Angeles, A.; Bueno, C.; Menéndez, P.; Martín-Subero, J.I.; Daley, G.Q.; Freije, J.M.; López-Otín, C. NF-κB activation impairs somatic cell reprogramming in ageing. Nat. Cell Biol. 2015, 17, 1004–1013. [Google Scholar] [CrossRef] [PubMed]
- Gerondakis, S.; Grumont, R.; Gugasyan, R.; Wong, L.; Isomura, I.; Ho, W.; Banerjee, A. Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic models. Oncogene 2006, 25, 6781–6799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tilstra, J.S.; Robinson, A.R.; Wang, J.; Gregg, S.Q.; Clauson, C.L.; Reay, D.P.; Nasto, L.A.; St Croix, C.M.; Usas, A.; Vo, N.; et al. NF-kappaB inhibition delays DNA damage-induced senescence and aging in mice. J. Clin. Investig. 2012, 122, 2601–2612. [Google Scholar] [CrossRef] [Green Version]
- Leung, T.H.; Zhang, L.F.; Wang, J.; Ning, S.; Knox, S.J.; Kim, S.K. Topical hypochlorite ameliorates NF-kappaB-mediated skin diseases in mice. J. Clin. Investig. 2013, 123, 5361–5370. [Google Scholar] [CrossRef] [Green Version]
- Acharyya, S.; Villalta, S.A.; Bakkar, N.; Bupha-Intr, T.; Janssen, P.M.; Carathers, M.; Li, Z.W.; Beg, A.A.; Ghosh, S.; Sahenk, Z.; et al. Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy. J. Clin. Investig. 2007, 117, 889–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, G.; Li, J.; Purkayastha, S.; Tang, Y.; Zhang, H.; Yin, Y.; Li, B.; Liu, G.; Cai, D. Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH. Nature 2013, 497, 211–216. [Google Scholar] [CrossRef] [PubMed]
- Villeda, S.A.; Luo, J.; Mosher, K.I.; Zou, B.; Britschgi, M.; Bieri, G.; Stan, T.M.; Fainberg, N.; Ding, Z.; Eggel, A.; et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011, 477, 90–94. [Google Scholar] [CrossRef] [Green Version]
- Smith, L.K.; He, Y.; Park, J.S.; Bieri, G.; Snethlage, C.E.; Lin, K.; Gontier, G.; Wabl, R.; Plambeck, K.E.; Udeochu, J.; et al. β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat. Med. 2015, 21, 932–937. [Google Scholar] [CrossRef] [Green Version]
- Chung, H.Y.; Cesari, M.; Anton, S.; Marzetti, E.; Giovannini, S.; Seo, A.Y.; Carter, C.; Yu, B.P.; Leeuwenburgh, C. Molecular inflammation: Underpinnings of aging and age-related diseases. Ageing Res. Rev. 2009, 8, 18–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xia, S.; Zhang, X.; Zheng, S.; Khanabdali, R.; Kalionis, B.; Wu, J.; Wan, W.; Tai, X. An Update on Inflamm-Aging: Mechanisms, Prevention, and Treatment. J. Immunol. Res. 2016, 2016, 8426874. [Google Scholar] [CrossRef]
- Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
- Balistreri, C.R.; Candore, G.; Accardi, G.; Colonna-Romano, G.; Lio, D. NF-kappaB pathway activators as potential ageing biomarkers: Targets for new therapeutic strategies. Immun. Ageing I A 2013, 10, 24. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Perkins, N.D. Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat. Rev. Mol. Cell Biol. 2007, 8, 49–62. [Google Scholar] [CrossRef] [PubMed]
- Fletcher, E.L.; Jobling, A.I.; Greferath, U.; Mills, S.A.; Waugh, M.; Ho, T.; de Iongh, R.U.; Phipps, J.A.; Vessey, K.A. Studying age-related macular degeneration using animal models. Optom. Vis. Sci. Off. Publ. Am. Acad. Optom. 2014, 91, 878–886. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Zeng, Q.; Barış, M.; Tezel, G. Transgenic inhibition of astroglial NF-κB restrains the neuroinflammatory and neurodegenerative outcomes of experimental mouse glaucoma. J. Neuroinflamm. 2020, 17, 252. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Asbell, P.A. The core mechanism of dry eye disease is inflammation. Eye Contact Lens 2014, 40, 248–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, J.; Jimi, E.; Zeiss, C.; Hayden, M.S.; Ghosh, S. Constitutively active NF-kappaB triggers systemic TNFalpha-dependent inflammation and localized TNFalpha-independent inflammatory disease. Genes Dev. 2010, 24, 1709–1717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellucci, A.; Bubacco, L.; Longhena, F.; Parrella, E.; Faustini, G.; Porrini, V.; Bono, F.; Missale, C.; Pizzi, M. Nuclear Factor-κB Dysregulation and α-Synuclein Pathology: Critical Interplay in the Pathogenesis of Parkinson’s Disease. Front. Aging Neurosci. 2020, 12, 68. [Google Scholar] [CrossRef] [Green Version]
- Camandola, S.; Mattson, M.P. NF-kappa B as a therapeutic target in neurodegenerative diseases. Expert Opin. Ther. Targets 2007, 11, 123–132. [Google Scholar] [CrossRef] [PubMed]
- Parrella, E.; Bellucci, A.; Porrini, V.; Benarese, M.; Lanzillotta, A.; Faustini, G.; Longhena, F.; Abate, G.; Uberti, D.; Pizzi, M. NF-κB/c-Rel deficiency causes Parkinson’s disease-like prodromal symptoms and progressive pathology in mice. Transl. Neurodegener. 2019, 8, 16. [Google Scholar] [CrossRef] [PubMed]
- Baiguera, C.; Alghisi, M.; Pinna, A.; Bellucci, A.; De Luca, M.A.; Frau, L.; Morelli, M.; Ingrassia, R.; Benarese, M.; Porrini, V.; et al. Late-onset Parkinsonism in NFκB/c-Rel-deficient mice. Brain A J. Neurol. 2012, 135, 2750–2765. [Google Scholar] [CrossRef] [Green Version]
- Jha, N.K.; Jha, S.K.; Kar, R.; Nand, P.; Swati, K.; Goswami, V.K. Nuclear factor-kappa β as a therapeutic target for Alzheimer’s disease. J. Neurochem. 2019, 150, 113–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, S.V.; Kounatidis, I. Nuclear Factor-Kappa B and Alzheimer Disease, Unifying Genetic and Environmental Risk Factors from Cell to Humans. Front. Immunol. 2017, 8, 1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Terai, K.; Matsuo, A.; McGeer, P.L. Enhancement of immunoreactivity for NF-kappa B in the hippocampal formation and cerebral cortex of Alzheimer’s disease. Brain Res. 1996, 735, 159–168. [Google Scholar] [CrossRef]
- Kaltschmidt, B.; Uherek, M.; Volk, B.; Baeuerle, P.A.; Kaltschmidt, C. Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proc. Natl. Acad. Sci. USA 1997, 94, 2642–2647. [Google Scholar] [CrossRef] [Green Version]
- Lian, H.; Yang, L.; Cole, A.; Sun, L.; Chiang, A.C.; Fowler, S.W.; Shim, D.J.; Rodriguez-Rivera, J.; Taglialatela, G.; Jankowsky, J.L.; et al. NFκB-activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer’s disease. Neuron 2015, 85, 101–115. [Google Scholar] [CrossRef] [Green Version]
- Kiernan, M.C.; Vucic, S.; Cheah, B.C.; Turner, M.R.; Eisen, A.; Hardiman, O.; Burrell, J.R.; Zoing, M.C. Amyotrophic lateral sclerosis. Lancet 2011, 377, 942–955. [Google Scholar] [CrossRef] [Green Version]
- Picher-Martel, V.; Dutta, K.; Phaneuf, D.; Sobue, G.; Julien, J.P. Ubiquilin-2 drives NF-κB activity and cytosolic TDP-43 aggregation in neuronal cells. Mol. Brain 2015, 8, 71. [Google Scholar] [CrossRef] [Green Version]
- Dutta, K.; Thammisetty, S.S.; Boutej, H.; Bareil, C.; Julien, J.P. Mitigation of ALS Pathology by Neuron-Specific Inhibition of Nuclear Factor Kappa B Signaling. J. Neurosci. 2020, 40, 5137–5154. [Google Scholar] [CrossRef] [PubMed]
- Sacitharan, P.K. Ageing and Osteoarthritis. Subcell. Biochem. 2019, 91, 123–159. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, N.D.; Ahlborg, H.G.; Center, J.R.; Eisman, J.A.; Nguyen, T.V. Residual lifetime risk of fractures in women and men. J. Bone Miner. Res. 2007, 22, 781–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abu-Amer, Y. NF-κB signaling and bone resorption. Osteoporos. Int. 2013, 24, 2377–2386. [Google Scholar] [CrossRef] [Green Version]
- Ginaldi, L.; Di Benedetto, M.C.; De Martinis, M. Osteoporosis, inflammation and ageing. Immun. Ageing I A 2005, 2, 14. [Google Scholar] [CrossRef] [Green Version]
- Chang, J.; Wang, Z.; Tang, E.; Fan, Z.; McCauley, L.; Franceschi, R.; Guan, K.; Krebsbach, P.H.; Wang, C.Y. Inhibition of osteoblastic bone formation by nuclear factor-kappaB. Nat. Med. 2009, 15, 682–689. [Google Scholar] [CrossRef]
- Lin, T.; Pajarinen, J.; Kohno, Y.; Nabeshima, A.; Lu, L.; Nathan, K.; Yao, Z.; Wu, J.Y.; Goodman, S. Increased NF-kB activity in osteoprogenitor-lineage cells impairs the balance of bone versus fat in the marrow of skeletally mature mice. Regen. Eng. Transl. Med. 2020, 6, 69–77. [Google Scholar] [CrossRef]
- Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtahl, A.J.; Pelletier, J.P. Osteoarthritis. Nat. Rev. Dis. Prim. 2016, 2, 16072. [Google Scholar] [CrossRef] [Green Version]
- Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
- Olivotto, E.; Otero, M.; Astolfi, A.; Platano, D.; Facchini, A.; Pagani, S.; Flamigni, F.; Facchini, A.; Goldring, M.B.; Borzì, R.M.; et al. IKKα/CHUK regulates extracellular matrix remodeling independent of its kinase activity to facilitate articular chondrocyte differentiation. PLoS ONE 2013, 8, e73024. [Google Scholar] [CrossRef]
- Culley, K.L.; Lessard, S.G.; Green, J.D.; Quinn, J.; Chang, J.; Khilnani, T.; Wondimu, E.B.; Dragomir, C.L.; Marcu, K.B.; Goldring, M.B.; et al. Inducible knockout of CHUK/IKKα in adult chondrocytes reduces progression of cartilage degradation in a surgical model of osteoarthritis. Sci. Rep. 2019, 9, 8905. [Google Scholar] [CrossRef] [Green Version]
- Yeung, S.S.Y.; Reijnierse, E.M.; Pham, V.K.; Trappenburg, M.C.; Lim, W.K.; Meskers, C.G.M.; Maier, A.B. Sarcopenia and its association with falls and fractures in older adults: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2019, 10, 485–500. [Google Scholar] [CrossRef] [Green Version]
- Shou, J.; Chen, P.J.; Xiao, W.H. Mechanism of increased risk of insulin resistance in aging skeletal muscle. Diabetol. Metab. Syndr. 2020, 12, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tisdale, M.J. Cachexia in cancer patients. Nat. Rev. Cancer 2002, 2, 862–871. [Google Scholar] [CrossRef] [PubMed]
- He, W.A.; Berardi, E.; Cardillo, V.M.; Acharyya, S.; Aulino, P.; Thomas-Ahner, J.; Wang, J.; Bloomston, M.; Muscarella, P.; Nau, P.; et al. NF-κB-mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J. Clin. Investig. 2013, 123, 4821–4835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hunter, R.B.; Kandarian, S.C. Disruption of either the Nfkb1 or the Bcl3 gene inhibits skeletal muscle atrophy. J. Clin. Investig. 2004, 114, 1504–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mourkioti, F.; Kratsios, P.; Luedde, T.; Song, Y.H.; Delafontaine, P.; Adami, R.; Parente, V.; Bottinelli, R.; Pasparakis, M.; Rosenthal, N. Targeted ablation of IKK2 improves skeletal muscle strength, maintains mass, and promotes regeneration. J. Clin. Investig. 2006, 116, 2945–2954. [Google Scholar] [CrossRef] [Green Version]
- Judge, A.R.; Koncarevic, A.; Hunter, R.B.; Liou, H.C.; Jackman, R.W.; Kandarian, S.C. Role for IkappaBalpha, but not c-Rel, in skeletal muscle atrophy. Am. J. Physiol. Cell Physiol. 2007, 292, C372–C382. [Google Scholar] [CrossRef] [Green Version]
- Van Gammeren, D.; Damrauer, J.S.; Jackman, R.W.; Kandarian, S.C. The IkappaB kinases IKKalpha and IKKbeta are necessary and sufficient for skeletal muscle atrophy. FASEB J. 2009, 23, 362–370. [Google Scholar] [CrossRef] [PubMed]
- Lakatta, E.G. Arterial and cardiac aging: Major shareholders in cardiovascular disease enterprises: Part III: Cellular and molecular clues to heart and arterial aging. Circulation 2003, 107, 490–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ungvari, Z.; Csiszar, A.; Kaley, G. Vascular inflammation in aging. Herz 2004, 29, 733–740. [Google Scholar] [CrossRef] [PubMed]
- Dick, S.A.; Epelman, S. Chronic Heart Failure and Inflammation: What Do We Really Know? Circ. Res. 2016, 119, 159–176. [Google Scholar] [CrossRef] [Green Version]
- Fiordelisi, A.; Iaccarino, G.; Morisco, C.; Coscioni, E.; Sorriento, D. NFkappaB is a Key Player in the Crosstalk between Inflammation and Cardiovascular Diseases. Int. J. Mol. Sci. 2019, 20, 1599. [Google Scholar] [CrossRef] [Green Version]
- Csiszar, A.; Wang, M.; Lakatta, E.G.; Ungvari, Z. Inflammation and endothelial dysfunction during aging: Role of NF-kappaB. J. Appl. Physiol. 2008, 105, 1333–1341. [Google Scholar] [CrossRef] [Green Version]
- Van der Heiden, K.; Cuhlmann, S.; Luong le, A.; Zakkar, M.; Evans, P.C. Role of nuclear factor kappaB in cardiovascular health and disease. Clin. Sci. 2010, 118, 593–605. [Google Scholar] [CrossRef]
- Mann, D.L. Inflammatory mediators and the failing heart: Past, present, and the foreseeable future. Circ. Res. 2002, 91, 988–998. [Google Scholar] [CrossRef] [PubMed]
- Purcell, N.H.; Molkentin, J.D. Is nuclear factor kappaB an attractive therapeutic target for treating cardiac hypertrophy? Circulation 2003, 108, 638–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwasaki, A.; Matsumori, A.; Yamada, T.; Shioi, T.; Wang, W.; Ono, K.; Nishio, R.; Okada, M.; Sasayama, S. Pimobendan inhibits the production of proinflammatory cytokines and gene expression of inducible nitric oxide synthase in a murine model of viral myocarditis. J. Am. Coll. Cardiol. 1999, 33, 1400–1407. [Google Scholar] [CrossRef] [Green Version]
- Matsumori, A.; Nunokawa, Y.; Sasayama, S. Pimobendan inhibits the activation of transcription factor NF-kappaB: A mechanism which explains its inhibition of cytokine production and inducible nitric oxide synthase. Life Sci. 2000, 67, 2513–2519. [Google Scholar] [CrossRef]
- Kawano, S.; Kubota, T.; Monden, Y.; Kawamura, N.; Tsutsui, H.; Takeshita, A.; Sunagawa, K. Blockade of NF-kappaB ameliorates myocardial hypertrophy in response to chronic infusion of angiotensin II. Cardiovasc. Res. 2005, 67, 689–698. [Google Scholar] [CrossRef]
- Frantz, S.; Hu, K.; Bayer, B.; Gerondakis, S.; Strotmann, J.; Adamek, A.; Ertl, G.; Bauersachs, J. Absence of NF-kappaB subunit p50 improves heart failure after myocardial infarction. FASEB J. 2006, 20, 1918–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawano, S.; Kubota, T.; Monden, Y.; Tsutsumi, T.; Inoue, T.; Kawamura, N.; Tsutsui, H.; Sunagawa, K. Blockade of NF-kappaB improves cardiac function and survival after myocardial infarction. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H1337–H1344. [Google Scholar] [CrossRef] [Green Version]
- Kawamura, N.; Kubota, T.; Kawano, S.; Monden, Y.; Feldman, A.M.; Tsutsui, H.; Takeshita, A.; Sunagawa, K. Blockade of NF-kappaB improves cardiac function and survival without affecting inflammation in TNF-alpha-induced cardiomyopathy. Cardiovasc. Res. 2005, 66, 520–529. [Google Scholar] [CrossRef]
- Maier, H.J.; Schips, T.G.; Wietelmann, A.; Krüger, M.; Brunner, C.; Sauter, M.; Klingel, K.; Böttger, T.; Braun, T.; Wirth, T. Cardiomyocyte-specific IκB kinase (IKK)/NF-κB activation induces reversible inflammatory cardiomyopathy and heart failure. Proc. Natl. Acad. Sci. USA 2012, 109, 11794–11799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamid, T.; Guo, S.Z.; Kingery, J.R.; Xiang, X.; Dawn, B.; Prabhu, S.D. Cardiomyocyte NF-κB p65 promotes adverse remodelling, apoptosis, and endoplasmic reticulum stress in heart failure. Cardiovasc. Res. 2011, 89, 129–138. [Google Scholar] [CrossRef]
- Liu, Q.; Chen, Y.; Auger-Messier, M.; Molkentin, J.D. Interaction between NFκB and NFAT coordinates cardiac hypertrophy and pathological remodeling. Circ. Res. 2012, 110, 1077–1086. [Google Scholar] [CrossRef] [Green Version]
- Emini Veseli, B.; Perrotta, P.; De Meyer, G.R.A.; Roth, L.; Van der Donckt, C.; Martinet, W.; De Meyer, G.R.Y. Animal models of atherosclerosis. Eur. J. Pharmacol. 2017, 816, 3–13. [Google Scholar] [CrossRef]
- Gareus, R.; Kotsaki, E.; Xanthoulea, S.; van der Made, I.; Gijbels, M.J.; Kardakaris, R.; Polykratis, A.; Kollias, G.; de Winther, M.P.; Pasparakis, M. Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis. Cell Metab. 2008, 8, 372–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, D.; Fang, G.; Mao, S.Z.; Ye, X.; Liu, G.; Gong, Y.; Liu, S.F. Chronic intermittent hypoxia induces atherosclerosis by NF-κB-dependent mechanisms. Biochim. Biophys. Acta 2012, 1822, 1650–1659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, G.; Song, D.; Ye, X.; Mao, S.Z.; Liu, G.; Liu, S.F. Chronic intermittent hypoxia exposure induces atherosclerosis in ApoE knockout mice: Role of NF-κB p50. Am. J. Pathol. 2012, 181, 1530–1539. [Google Scholar] [CrossRef] [PubMed]
- Ryan, S.; Taylor, C.T.; McNicholas, W.T. Selective activation of inflammatory pathways by intermittent hypoxia in obstructive sleep apnea syndrome. Circulation 2005, 112, 2660–2667. [Google Scholar] [CrossRef] [Green Version]
- Fletcher, E.C. Invited review: Physiological consequences of intermittent hypoxia: Systemic blood pressure. J. Appl. Physiol. (Bethesda, Md. 1985) 2001, 90, 1600–1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drager, L.F.; Polotsky, V.Y.; Lorenzi-Filho, G. Obstructive sleep apnea: An emerging risk factor for atherosclerosis. Chest 2011, 140, 534–542. [Google Scholar] [CrossRef] [Green Version]
- Sui, Y.; Park, S.H.; Xu, J.; Monette, S.; Helsley, R.N.; Han, S.S.; Zhou, C. IKKβ links vascular inflammation to obesity and atherosclerosis. J. Exp. Med. 2014, 211, 869–886. [Google Scholar] [CrossRef]
- Dandona, P.; Aljada, A.; Bandyopadhyay, A. Inflammation: The link between insulin resistance, obesity and diabetes. Trends Immunol. 2004, 25, 4–7. [Google Scholar] [CrossRef] [PubMed]
- Avgerinos, K.I.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metab Clin. Exp. 2019, 92, 121–135. [Google Scholar] [CrossRef] [PubMed]
- Yuan, M.; Konstantopoulos, N.; Lee, J.; Hansen, L.; Li, Z.W.; Karin, M.; Shoelson, S.E. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science 2001, 293, 1673–1677. [Google Scholar] [CrossRef]
- Cai, D.; Yuan, M.; Frantz, D.F.; Melendez, P.A.; Hansen, L.; Lee, J.; Shoelson, S.E. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat. Med. 2005, 11, 183–190. [Google Scholar] [CrossRef]
- Arkan, M.C.; Hevener, A.L.; Greten, F.R.; Maeda, S.; Li, Z.W.; Long, J.M.; Wynshaw-Boris, A.; Poli, G.; Olefsky, J.; Karin, M. IKK-beta links inflammation to obesity-induced insulin resistance. Nat. Med. 2005, 11, 191–198. [Google Scholar] [CrossRef]
- Ke, B.; Zhao, Z.; Ye, X.; Gao, Z.; Manganiello, V.; Wu, B.; Ye, J. Inactivation of NF-κB p65 (RelA) in Liver Improves Insulin Sensitivity and Inhibits cAMP/PKA Pathway. Diabetes 2015, 64, 3355–3362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasegawa, Y.; Saito, T.; Ogihara, T.; Ishigaki, Y.; Yamada, T.; Imai, J.; Uno, K.; Gao, J.; Kaneko, K.; Shimosawa, T.; et al. Blockade of the nuclear factor-κB pathway in the endothelium prevents insulin resistance and prolongs life spans. Circulation 2012, 125, 1122–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, T.; Zhang, J.; Yin, J.; Staszkiewicz, J.; Gawronska-Kozak, B.; Jung, D.Y.; Ko, H.J.; Ong, H.; Kim, J.K.; Mynatt, R.; et al. Uncoupling of inflammation and insulin resistance by NF-kappaB in transgenic mice through elevated energy expenditure. J. Biol. Chem. 2010, 285, 4637–4644. [Google Scholar] [CrossRef] [Green Version]
- Gao, Z.; Yin, J.; Zhang, J.; He, Q.; McGuinness, O.P.; Ye, J. Inactivation of NF-kappaB p50 leads to insulin sensitization in liver through post-translational inhibition of p70S6K. J. Biol. Chem. 2009, 284, 18368–18376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minegishi, Y.; Haramizu, S.; Misawa, K.; Shimotoyodome, A.; Hase, T.; Murase, T. Deletion of nuclear factor-κB p50 upregulates fatty acid utilization and contributes to an anti-obesity and high-endurance phenotype in mice. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E523–E533. [Google Scholar] [CrossRef] [Green Version]
- Bhatt, B.A.; Dedousis, N.; Sipula, I.J.; O’Doherty, R.M. Elevated metabolic rate and skeletal muscle oxidative metabolism contribute to the reduced susceptibility of NF-κB p50 null mice to obesity. Physiol. Rep. 2018, 6, e13836. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Zhang, X.; Zhao, Z.; Lu, H.; Ke, B.; Ye, X.; Wu, B.; Ye, J. NF-κ B/HDAC1/SREBP1c pathway mediates the inflammation signal in progression of hepatic steatosis. Acta Pharm. Sin. B 2020, 10, 825–836. [Google Scholar] [CrossRef] [PubMed]
- Fausto, N. Mouse liver tumorigenesis: Models, mechanisms, and relevance to human disease. Semin. Liver Dis. 1999, 19, 243–252. [Google Scholar] [CrossRef]
- Maeda, S.; Kamata, H.; Luo, J.L.; Leffert, H.; Karin, M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005, 121, 977–990. [Google Scholar] [CrossRef] [Green Version]
- Naugler, W.E.; Sakurai, T.; Kim, S.; Maeda, S.; Kim, K.; Elsharkawy, A.M.; Karin, M. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007, 317, 121–124. [Google Scholar] [CrossRef] [Green Version]
- Wilson, C.L.; Jurk, D.; Fullard, N.; Banks, P.; Page, A.; Luli, S.; Elsharkawy, A.M.; Gieling, R.G.; Chakraborty, J.B.; Fox, C.; et al. NFκB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat. Commun. 2015, 6, 6818. [Google Scholar] [CrossRef] [Green Version]
- Inaba, Y.; Furutani, T.; Kimura, K.; Watanabe, H.; Haga, S.; Kido, Y.; Matsumoto, M.; Yamamoto, Y.; Harada, K.; Kaneko, S.; et al. Growth arrest and DNA damage-inducible 34 regulates liver regeneration in hepatic steatosis in mice. Hepatology 2015, 61, 1343–1356. [Google Scholar] [CrossRef]
- Michalopoulos, G.K. Principles of liver regeneration and growth homeostasis. Compr. Physiol. 2013, 3, 485–513. [Google Scholar] [CrossRef]
- Richardson, M.M.; Jonsson, J.R.; Powell, E.E.; Brunt, E.M.; Neuschwander-Tetri, B.A.; Bhathal, P.S.; Dixon, J.B.; Weltman, M.D.; Tilg, H.; Moschen, A.R.; et al. Progressive fibrosis in nonalcoholic steatohepatitis: Association with altered regeneration and a ductular reaction. Gastroenterology 2007, 133, 80–90. [Google Scholar] [CrossRef]
- Sancho-Bru, P.; Altamirano, J.; Rodrigo-Torres, D.; Coll, M.; Millán, C.; José Lozano, J.; Miquel, R.; Arroyo, V.; Caballería, J.; Ginès, P.; et al. Liver progenitor cell markers correlate with liver damage and predict short-term mortality in patients with alcoholic hepatitis. Hepatology 2012, 55, 1931–1941. [Google Scholar] [CrossRef]
- Xiong, Y.; Torsoni, A.S.; Wu, F.; Shen, H.; Liu, Y.; Zhong, X.; Canet, M.J.; Shah, Y.M.; Omary, M.B.; Liu, Y.; et al. Hepatic NF-kB-inducing kinase (NIK) suppresses mouse liver regeneration in acute and chronic liver diseases. eLife 2018, 7, e34152. [Google Scholar] [CrossRef] [PubMed]
- Shen, H.; Sheng, L.; Chen, Z.; Jiang, L.; Su, H.; Yin, L.; Omary, M.B.; Rui, L. Mouse hepatocyte overexpression of NF-κB-inducing kinase (NIK) triggers fatal macrophage-dependent liver injury and fibrosis. Hepatology 2014, 60, 2065–2076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prabhakar, M.; Ershler, W.B.; Longo, D.L. Bone marrow, thymus and blood: Changes across the lifespan. Ageing Health 2009, 5, 385–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlson, M.E.; Conboy, I.M. Loss of stem cell regenerative capacity within aged niches. Aging Cell 2007, 6, 371–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lavasani, M.; Robinson, A.R.; Lu, A.; Song, M.; Feduska, J.M.; Ahani, B.; Tilstra, J.S.; Feldman, C.H.; Robbins, P.D.; Niedernhofer, L.J.; et al. Muscle-derived stem/progenitor cell dysfunction limits healthspan and lifespan in a murine progeria model. Nat. Commun. 2012, 3, 608. [Google Scholar] [CrossRef] [Green Version]
- Wright, V.; Peng, H.; Usas, A.; Young, B.; Gearhart, B.; Cummins, J.; Huard, J. BMP4-expressing muscle-derived stem cells differentiate into osteogenic lineage and improve bone healing in immunocompetent mice. Mol. Ther. 2002, 6, 169–178. [Google Scholar] [CrossRef]
- Kuroda, R.; Usas, A.; Kubo, S.; Corsi, K.; Peng, H.; Rose, T.; Cummins, J.; Fu, F.H.; Huard, J. Cartilage repair using bone morphogenetic protein 4 and muscle-derived stem cells. Arthritis Rheumatol. 2006, 54, 433–442. [Google Scholar] [CrossRef] [PubMed]
- Proto, J.D.; Lu, A.; Dorronsoro, A.; Scibetta, A.; Robbins, P.D.; Niedernhofer, L.J.; Huard, J. Inhibition of NF-κB improves the stress resistance and myogenic differentiation of MDSPCs isolated from naturally aged mice. PLoS ONE 2017, 12, e0179270. [Google Scholar] [CrossRef] [Green Version]
- Tichy, E.D.; Ma, N.; Sidibe, D.; Loro, E.; Kocan, J.; Chen, D.Z.; Khurana, T.S.; Hasty, P.; Mourkioti, F. Persistent NF-κB activation in muscle stem cells induces proliferation-independent telomere shortening. Cell Rep. 2021, 35, 109098. [Google Scholar] [CrossRef] [PubMed]
- Prescott, J.A.; Cook, S.J. Targeting IKKβ in Cancer: Challenges and Opportunities for the Therapeutic Utilisation of IKKβ Inhibitors. Cells 2018, 7, 115. [Google Scholar] [CrossRef] [Green Version]
- Ramadass, V.; Vaiyapuri, T.; Tergaonkar, V. Small Molecule NF-κB Pathway Inhibitors in Clinic. Int. J. Mol. Sci. 2020, 21, 5164. [Google Scholar] [CrossRef] [PubMed]
NF-κB Gene | Genetic Modification | Aging Phenotype and Proposed Mechanisms | Cancer Development |
---|---|---|---|
Nfkb1−/− [65] | Nfkb1−/− mice, lacking both the p105 precursor and p50 subunit | Premature ageing. Reduced regeneration in liver and gut. Chronic inflammation; ROS-mediated exacerbation of telomere dysfunction and cell senescence. Mice between 12 and 132 weeks of age were analyzed. | |
Nfkb1−/− [64] | Nfkb1−/− mice, | Increase inflammation, premature cellular senescence, phospho-H2AX accumulation and lower levels of spontaneous apoptosis in brains and MEFs from Nfkb1−/−, compared to Nfkb1+/+ mice. Mice of 12 and 18 months of age were analyzed. | |
Cyld [66] | K5-CyldC/S mice expressing a dominant negative, catalytically inactive, form of CYLD driven by the K5 promoter. | Premature ageing of skin, pancreas, liver, stomach, and thymus. Chronic over-activation of NF-κB; systemic inflammation; VEGF and c-Myc induction. Spontaneous development of different types of tumors. Young (1- to 9-month-old) and old (20- to 24-month-old) mice were analyzed. | Lung, gastric, non-melanoma skin cancer; HCC. |
Ikkβ [67] | K5-Ikkβ mice overexpressing IKKβ under the control of the K5 promoter. | Over-activation of the canonical pathway of NF-κB; chronic inflammation in oral epithelia. Unpublished data: skin atrophy, loss of hair follicles and fat and sebaceous gland hyperplasia at early age (6 months). Mice between 3 and 20 months of age were analyzed. | Spontaneous tumors in oral epithelia. |
Rela−/− [46] | Sirt6/rela+/− mice. Derived from Sirt6-deficient mice crossed with Rela+/− mice. | Deficiency of mammalian SIRT6 leads to shortened lifespan and an ageing-like phenotype. SIRT6 interacts with p65 attenuating NF-κB signaling. Multiple Sirt6-deficient tissues show increased activity NF-κB -driven gene expression programs. Haploinsufficiency of rela rescues the early lethality and degenerative syndrome of Sirt6-deficient mice. 21- to 29-day-old mice were analyzed. | |
Rela−/− [74] | A model of XFE progeroid syndrome, Ercc1–/Δ mice; murine Ercc1–/– mice: deficient in DNA excision repair protein ERCC-1. Crosses with Rela+/– mice. | Ercc1–/Δ mice present a progeroid phenotype. Genetic depletion of one allele of the Rela gene or treatment with a pharmacological inhibitor of IKK, delayed the age-related symptoms and pathologies in Ercc1–/Δ mice such as trembling, kyphosis, and sarcopenia, as well as oxidative DNA damage and stress, and cellular senescence. Mice of 21 days of age, as well as 3- and 24-month-old mice were studied. | |
Rela−/− [68] | Zmspte24−/−Rela+/− mice derived from Zmpste24-deficient mice crossed with rela+/− mice. | Progeroid syndrome: Progeroid laminopathy by defects of the nuclear lamina, phenocopying most features of HGPS (Gilford Progeria Syndrome). Zmpste24−/− mice accumulate farnesylated prelamin A at the nuclear envelope, leading to activation of ATM kinase, which cooperates with nuclear IKKγ. This results in the activation of NF-κB and up-regulation of proinflammatory cytokines. Chronic inflammatory state affects distant cells. Zmpste24−/−Rela+/− mice showed recovery of skin and immunological alterations and display a retardation in the ageing process and extended longevity. Three-month-old mice were analyzed. | |
Ikkβ [53] | MIKK mice with muscle-specific expression of constitutively active IkkβSS177/181EE (muscle creatine kinase promoter, MCK). MISR mice: expression of IkBα SS32/36AA (superrepressor) in muscle that inactivates NF-κB. | MIKK mice shows constitutive activation of NF-κB and undergo decreased skeletal muscle mass and body weight; reduced contractile function in muscles. MIKK x MISR mice reversed muscle atrophy. IKKβ/NF-κB activation causes accelerated protein breakdown through ubiquitin-dependent proteolysis mediated by E3ligase MuRF1. Pharmacologic NF-κB inhibition in MIKK mice reversed the muscle atrophy. Mice between one week and six months of age were analyzed. | |
Ikkβ,Rela [76] | Ablation of 1 allele of Rela in a mdx mouse model of DMD (dystrophin-deficient mice) and conditional deletion of Ikkβ in these mice. | DMD mice resembles a premature aging syndrome: their myogenic stem cells become prematurely senescent. The deletion of 1 allele of rela improved the pathology of dystrophic muscles in mdx mice. Ikkβ deletion both in the immune compartment and skeletal muscle fibers reduces inflammation and necrosis through the reduction of NF-κB activity and promotes regeneration in dystrophic muscles. Mice between 4- to -12 weeks of age were analyzed. | |
Ikkβ [77] | N/Ikkβlox/lox mice: Ikkβlox/lox mice crossed with Nestin-Cre mice. | N/Ikkβlox/lox mice showed a pronounced phenotype of longevity, with median lifespan 23% longer. The longevity phenotype of this genetic model could be a result of IKKβ inhibition in neurons and glia. IKK-β and NF-κB repress gonadotropin-releasing hormone (GnRH) to mediate aging-related hypothalamic GnRH decline associated to systemic ageing. Mice aged 6 to 26 weeks were studied. |
NF-κB Gene | Genetic Modification | Recapitulated Disease and Proposed Mechanisms |
---|---|---|
Eye Diseases | ||
Ikkβ [87] | Specific knock-out of Ikkβ in astroglia. Use of the cre/loxP system under the regulation of the mouse glial Gfap to generate the GFAP-ikkβ mice. | Glaucoma GFAP-ikkβ mice lacking Ikkβ in astroglia show a high decrease in ocular hypertension and impeded inflammatory and neurodegenerative outcomes in the retina and optic nerve. This is the result of the inhibition of astroglial NF-κB activation, and decreased levels of proinflammatory cytokines in astroglia. Two- to three-month-old mice were analyzed. |
Rela [89] | Generation of knock-in mice expressing the p65 S276D phosphomimetic protein, (p65PD mice), which show hyperactivation of NF-κB. Crossing of p65PD mice with mice deficient in Tnf receptor 1) to obtain the p65PD/Tnfrsf1a−/− mice. | Dry eye disease p65PD mice show a systemic hyperinflammatory condition due to increased TNF-α production, and died within 20 d after birth (5- and 10-day-old mice were studied). Crossing with Tnfr1 knockout mice rescues the phenotype, but aged p65PD/Tnfrsf1a−/− mice (3.5 to 8 months of age mice were analyzed) develop chronic keratitis or keratoconjunctivitis sicca (DED), with severe corneal epithelial lesions, marked neovascularization and inflammation of the corneal stroma, and increased levels of TNF-α, IL-1β, and MMP-9. DED chronic keratitis is independent of TNFR1 signaling but dependent on NF-κB. |
Neurodegenerative Diseases | ||
c-Rel [93] | c-Rel gene null mutation generated by inserting the neomycin cassette into the fifth exon of the c-Rel gene. | Parkinson’s disease c-Rel KO mice develop PD-like neuropathology with ageing, manifested by age-dependent locomotor deficits reminiscent of bradykinesia and muscle rigidity. Loss of dopaminergic neurons, increased levels of iron and DMT1, and accumulation of aggregated α-synuclein with activation of microglial cells An altered balance between p65- and c-REL-mediated effects on neurons is proposed. Mice aged 2- to 18-month-old were analyzed. |
c-Rel [92] | c-Rel gene null mutation generated by inserting the neomycin cassette into the fifth exon of the c-Rel gene. | Parkinson’s disease In addition to the phenotype described ([80]), c-Rel−/− mice show symptoms and pathology peculiar of prodromal syndrome and a Braak-like stereotyped diffusion of synucleinopathy mimicking sporadic Parkinson´s disease. Potential involvement of changes in levels of proteins controlling mitochondrial homeostasis, ROS generation and scavenging are proposed. Mice from 2 to 18 months of age were analyzed. |
Iκbα [98] | NcKO mice: mice with CNS-specific Iκbα deletion (by crossingiIκbα flox with Nestin-Cre mice); CcKO and GcKO mice: mice with selective Iκbα deletion in neurons or in astrocytes, respectively (by crossing IkBa flox with CaMKIIa-Cre mice; or with GFAP-Cre mice). | Alzheimer´s disease This model links Amyloid β with synaptic defects and neuronal hyperexcitability though the sustained NF-κB activity in astrocytes, which results in the release of the complement protein C3 and the disruption of the dendritic morphology and network function characteristic of AD. Dysregulation of neuron-glia interaction through NF-κB/C3/C3aR signaling contribute to synaptic dysfunction in AD. Mice from 2 to 18 months of age were studied. |
Iκbα [101] | Neuron-specific expression of IkBα super-repressor (IκBα-SR) driven by NFH neurofilament H promoter. Crosses of IκB-SR mice with familial ALS-linked mutant Tdp-43 (G348C and A315T) and Sod1 (G93A) mice. | Amyotrophic lateral sclerosis Mitigation of ALS symptoms and extension of life span in double transgenic mice IκBα-SR/TDP-43 and IκBα-SR/SOD1G93A mice. The inhibition of NF-κB activity by the expression of IκBα-SR in neurons alleviates TDP-43 in neuropathology through an induction of autophagy. Reduction of misfolded SOD1 levels in and IκBα-SR/SOD1G93A mice. Mice from 4 to 16 months of age were studied. |
Osteoporosis | ||
Ikkβ [107] | Osteoprogenitor (OP)-specific, doxycycline-regulated Ikkβ-activated mouse model (iNF-κB/OP). These mice have activated NF-κB signaling in OP-lineage cells upon DOX withdrawal in skeletally mature mice. | iNF-κB/OP mice, with increased NF-κB activation in OP-lineage cells, showed reduced bone mineral density in the femurs and tibias and increased bone marrow fat, resembling human osteoporosis. The mechanisms involved are the decreased expression of osteogenic markers (Runx2 and osteocalcin) and the increased adipogenic markers (PPAR-γ and C/EBP) in mesenchymal stem cells (MSCs) of iNF-κB/OP mice. Mice of 12 and 36 weeks of age were analyzed. |
Osteoarthritis | ||
Ikkα [111] | Aggrecan-driven cartilage-specific Ikkα KO mice in chondrocytes. cKO mice are treated with tamoxifen for the specific induction. | Wilt type (WT) and cKO underwent destabilization of the medial meniscus (DMM) surgery to induce OA. cKO mice are protected from cartilage degradation after surgical DMM. cKO mice showed both reduced cartilage degradation and collagenase activity. Mice from 12 to 24 weeks of age were studied. |
Muscle Diseases | ||
Nfκb1 [116] | Nfκb1−/− and bcl3−/− mice. | Mice with KO of Nfκb1 or Bcl3 gene are resistant to muscle protein loss and functional deficit due to muscular disuse; they exhibit inhibition of fiber atrophy. Activation of the canonical pathway of NF-κB by TNF-α is associated with muscle protein loss. |
Ikbα, c-Rel [118] | Expression of a dominant negative IκBα (IκBαΔN) in soleus muscles of rats (unloaded or weight bearing).c-Rel−/− mice are also used. | IκBαΔN expression in soleus muscles inhibits NF-κB activation and abolishes the unloading-induced increase in both NF-κB activation and total ubiquitinated protein, resulting in an inhibition of 40% fiber atrophy. The expression of genes upregulated in muscle atrophy is also inhibited (MAFbx/Atrogin-1, Nedd4, IEX, 4E-BP1, FOXO3a, Cathepsin L). |
Ikkβ [117] | IKKβmko mice exhibit muscle-specific NF-κB inhibition, through targeted deletion of Ikkβ. Muscle atrophy was induced by sciatic nerve denervation. | o mice, lacking NF-κB activation in muscle cells showed improved skeletal muscle strength and shifted muscle fiber distribution. In response to denervation, Ikkβ depletion protected against atrophy, maintaining fiber type, size, and strength, increasing protein synthesis (through the activation of protein synthetic pathways: AKT, GSK3α/β, mTOR, and p70S6K), and decreasing protein degradation (muscle RING finger 1 (MuRF1). Additionally, IKKβmko mice showed muscle regeneration after damage by enhanced satellite cell activation and reduced fibrosis. |
Ikkβ [115] | Mice with specific expression of a constitutive kinase-active Ikkβ in Pax7-expressing satellite cells (inducible activation by Tamoxifen). | The selective activation of IKKβ in Pax7+ cells of transgenic mice led to an aggravated decline in muscle mass and fiber size. Overexpression of Pax7, induced by serum factors from cachectic mice in an NF-κB–dependent manner, was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. |
NF-κB Gene | Genetic Modification | Age-Related Disease |
---|---|---|
Heart Diseases | ||
Nfkb1 [130] | Nfkb1-KO mice. Chronic infusion of angiotensin II, increases systemic blood pressure and pro-inflammatory cytokines in the myocardium and provokes ventricular hypertrophy. | Cardiac hypertrophy Nfkb1-deficiency attenuates myocardial inflammation and hypertrophy in response to chronic infusion of angiotensin II without deteriorating cardiac function. Blockade NF-κB activation leads to abrogation of JNK phosphorylation in response to chronic infusion of angiotensin II. Mice from 8- to 12-week-old were analyzed. |
Nfkb1 [133] | Nfkb1-KO mice. Cross of Nfkb1-KO mice with TNF-TG mice (with cardiac-specific overexpression of TNF-α) as a model of cytokine-induced cardiomyopathy. | Cardiac dysfunction and remodeling Blockade of NF-κB in Nfkb1-KO mice reverses cardiac dysfunction and remodeling; improves cardiac function and survival without affecting inflammation in TNF-α-induced cardiomyopathy; it also blocks MMP-9 activation. Neonatal to 12-week-old mice were studied. |
Nfkb1 [132] | Nfkb1-deficient mice. Myocardial infarction was induced by ligation of the left coronary artery. | Myocardial infarction Targeted disruption of Nfkb1 reduces ventricular rupture, and improves cardiac function and survival after myocardial infarction (MI) with amelioration of myocyte hypertrophy and interstitial fibrosis. Mechanisms proposed are: chronic inhibition of NF-κB; amelioration of myocyte hypertrophy and interstitial fibrosis; selective abrogation of JNK phosphorylation; lower infarction area. Mice from 8- to 26-week-old were analyzed. |
Nfkb1 [131] | Nfkb1-deficient mice. Myocardial infarction was induced by ligation of the left coronary artery. | Myocardial infarction Absence of the NF-κBp50 subunit improves early survival and reduces left ventricular dilatation after myocardial infarction. Absence of the NF-κBp50 subunit reduces collagen content and MMP-9 expression after myocardial infarction (which are involved in progressive left ventricular remodeling). Mice from 8- to 24 weeks of age were analyzed. |
Ikkβ, Iκbα, Nemo [134] | Tet O.IKKβ-CA mice. Inducible transgenic mice with cardiomyocyte-specific expression of constitutively active IKKβ. IκBα-3M; IKKβ-DN, mice with a floxed Nemo allele. For induction of viral myocarditis, mice were infected intraperitoneally with purified CVB3. | Cardiomyopathy and heart failure In adult animals, IKKβ activation led to inflammatory dilated cardiomyopathy and heart failure. Upon transgene inactivation, the disease was reversed, even at an advanced stage. In vivo expression of the IκBα superrepressor prevented the development of the disease. Constitutively active IKKβ expression induces myocyte atrophy; excessive inflammatory response with enhanced levels of inflammatory cytokines, and an IFN type I signature, with activation of the IFN-stimulated gene 15 (ISG15) pathway. In cardiomyocytes lacking Nemo, the induction of ISG15 was attenuated. Mice from 4- to 24-week-old were studied. |
Ikbα [135] | Tg-IκBαS32A,S36A mice with myocyte-specific expression of a transdominant mutant human IκBα (α-MyHC promoter) HF induced by surgery. | Heart failure Tg-IκBαS32A,S36A mice exhibit improved post-infarction survival and alleviated LV remodeling, They do not show pro-inflammatory cytokine expression or fibrosis. Display decreased apoptosis and stimulated adaptive endoplasmic reticulum stress responses in HF systolic function. Myocyte NF-κB abrogation induces a marked decrease of both NF-κB p65 activation and cytokine expression. Mice from 10- to 24-week-old were analyzed. |
Rela [136] | Cardiac-specific deletion of Rela using a Cre-loxP system. | Cardiac hypertrophy and remodeling Rela deficiency in the heart of mouse decreases the hypertrophic response after pressure overload stimulation, and the degree of pathological remodeling, while preserves contractile function. Transcriptional regulatory mechanism whereby NF-κB and NFAT directly interact and synergistically promote transcriptional activation in cardiomyocytes. 8- and 12-week-old mice were studied. |
Atherosclerosis | ||
Nfkb1 [139] | Nfkb1−/− mice, subjected to chronic intermittent hypoxia (CIH) and high cholesterol diet (HCD). | Nfkb1 gene deletion diminished CIH + HCD-induced NF-κB activation and abolished CIH + HCD induced atherosclerosis. Nfkb1 gene deletion through the blockage of NF-κB activation inhibits CIH + HCD-induced activation of three major atherogenic mechanisms: vascular wall inflammation, hypercholesterolemia (p50 reduces serum cholesterol level), and macrophage foam cell formation. Mice between 7- and 42.6-week-old were analyzed. |
Nfkb1 [140] | Apoe-KO mice and mice deficient in Nfkb1 gene (p50-KO) were crossed and obtained ApoE-p50-DKO mice. | CIH downregulates hepatic low-density lipoprotein receptor and HMG-CoA reductase expression in ApoE-p50-DKO but not in ApoE-KO mice, showing the protective role of p50 in CIH-induced atherosclerosis by inhibiting CIH-induced inflammation and hypercholesterolemia. 7- to 37-week-old were analyzed. |
Ikkγ, IkBα [138] | Endothelium-restricted inhibition of NF-κB by ablation of Nemo/IKKγ or expression of dominant-negative IkBα in ApoE-KO mice: Tie1NEMOEC-KO/ApoE−/−;Tie2NEMOEC-KO/ApoE−/− (this latter tamoxifen-induced); and Tie2DNIkBa/ApoE−/− mice were used. | IKKγ or IkBα ablation in endothelial cells of ApoE−/− mice (a well-established mouse model of atherosclerosis), reduced atherosclerotic plaque formation in mice fed with a cholesterol rich diet. Inhibition of NF-κB activation abrogates adhesion molecule induction in endothelial cells, impaired macrophage recruitment to atherosclerotic plaques, and reduced expression of cytokines and chemokines in the aorta. Mice between 8- and 18-week-old were analyzed |
Ikkβ [144] | Ikkβ deficiency in smooth muscle cells (SMCs) driven by a SM22Cre-Ikkβ-flox system. To increase susceptibility to spontaneous atherosclerotic lesion development, the SM22Cre+IkkβF/F mice were crossed with LDLR−/− mice. | Deficiency of IKKβ in SMCs rendered low density lipoprotein receptor-null mice resistant to vascular inflammation and atherosclerosis induced by high-fat feeding. SM22Cre+IkkBF/FLDLR−/− mice had decreased levels of inflammatory cytokines (IL-1β, TNF-α, and MCP-1) in the atherosclerotic lesions, arterial walls, and plasma, indicating a reduction in vascular inflammation associated with chronic inflammation. 16-week-old mice were analyzed. |
NF-κB Gene | Genetic Modification | Recapitulated Disease and Proposed Mechanisms |
---|---|---|
Type 2 Diabetes Mellitus/Obesity/Insulin Resistance | ||
Ikkβ [147] | Ikkβ+/− mice. Ikkβ+/+Lepob/ob obtained by crossing Ikkβ+/− mice with ob/ob mice (Lepob/ob), a mouse model for type 2 diabetes, obesity and insulin-resistance. | Ikkβ+/− mice are protected against the development of insulin resistance during high-fat feeding; they show lower concentrations of glucose and insulin in blood. Ikkβ heterozygosity also diminishes levels of blood glucose concentrations and improves insulin sensitivity in Lepob/ob mice. Eight-week-old mice were analyzed. |
Ikkβ [149] | Transgenic mice with conditional inhibition of Ikkβ in hepatocytes (Ikbkb∆hep) or myeloid cells (Ikbkb∆mye). | The inhibition of NF-κB pathway in myeloid cells improved insulin sensitivity in all tissues, while the inhibition of this pathway in hepatocytes maintained insulin sensitivity only in the liver: IKKβ acts locally in the liver, inducing proinflammatory mediators, which act only in a paracrine manner to downregulate insulin sensitivity in the liver, and systematically in myeloid cells. 3- to 14-month-old mice were studied. |
Ikkβ, Iκbα [148] | Constitutive activation of Ikkβ in hepatocytes (LIKK mice). Expression of Iκbα supper-repressor in the liver of mice (LISK mice). | LKK mice show a type 2 diabetes phenotype, with hyperglycemia, and hepatic and systemic diet-induced insulin resistance, through mechanisms involving hepatocellular activation of NF-κB; increased levels of cytokines in liver and muscle and activation of resident Kupffer cells. LISR mice reversed the phenotype of LIKK mice. Mice between 4 and 16 weeks of age were analyzed. |
Ikkβ [144] | Ikkβ deficiency in smooth muscle cells (SMCs) driven by a SM22Cre-Ikkβ-flox system. | Ikkβ-deficient mice were resistant to diet-induced obesity and metabolic disorders: lower glucose and insulin concentration in blood; reduced hepatic triglycerides and cholesterol. Mechanisms involved: inhibition of NF-κB; increased levels of the uncoupling protein-1 (UCP-1) and increased thermogenesis; lower expression of both hepatic lipogenic genes (SREBP1c, SCD-1, and PPARγ), and inflammatory cytokines. Mice from 8- to 28-week-old were analyzed. |
Rela [150] | Mice L-p65-KO with a deletion of Rela gene in the liver (p65+/− mice) were generated by crossing floxed-Rela and Alb-cre mice. | L-p65-KO exhibited improvement hepatic insulin sensitivity, and a decrease gene expression in hepatic gluconeogenesis (feed with a high fat diet). The inhibition of the NF-κB pathway decreased the expression levels of cAMP and the phosphorylation of CREB in the liver of L-p65-KO mice. The low levels of cAMP results from increased expression of PDE3B, a cAMP-degrading enzyme. Mice between 8 and 32 weeks of age were studied. |
Iκbα [151] | Mice expressing the Iκbα superrepressor under the Tie2 enhancer (E-DNIκB mice). Crossing of E-DNIκB mice (a genetic model for obesity-diabetes syndrome), generates the E-DNIkB; Ay/+ mice. | Functionally inhibition of NF-κB specifically in endothelial cells prevents obesity-related metabolic deteriorations, with a decrease both in the macrophage infiltration into adipose tissue, and plasma oxidative stress markers induced by obesity; it also prevents age-related insulin resistance and vascular senescence at the time that extents the life span. Mice aged 8 to 50 weeks were analyzed. |
Nfkb1 [153] | Nfkb1-KO mice. | Nfkb1-deficient mice have improved insulin sensitivity. They exhibit lower expression levels of the p70 ribosomal protein S6 kinase (p70s6k or S6K1) in the liver, and reduced activity of IKKβ and IKKγ. Mice from 6- to 26-week-old were analyzed. |
Nfkb1 [154] | Nfkb1-KO mice. | Nfkb1-KO mice had both higher fatty acid utilization and oxidation in the liver, and, when feeding with a high-fat diet, they are resistant to fat accumulation and adipose tissue inflammation. Mice between 1 and 23 weeks of age were studied. |
Nfkb1[155] | Nfkb1-KO mice. | Nfkb1-KO mice exhibits reduced body weight gain on a high-fat diet, reduced plasma triglyceride levels and adiposity. The reduced susceptibility to diet-induced obesity and dyslipidemia in p50-deficient mice results from an increase in metabolic rate, associated with elevated skeletal muscle oxidative metabolism and decreased DGAT2 expression. Mice between 10 and 34 weeks of age were analyzed. |
Liver Diseases | ||
Nfkb1 [160] | Nfkb1-deficient mice. Nfkb11S340A/S340A mice carrying a mutation designed to selectively disrupt p50:p50:HDAC1 complexes. | Chronic liver disease Loss of Nfkb1 promotes ageing associated chronic liver disease (CLD) (steatosis, neutrophilia, fibrosis, hepatocyte telomere damage, dysplasia) and HCC development. Hepatic lesions in aged Nfkb1-/- mice were associated with highly elevated numbers of hepatic neutrophils, which stimulates hepatocellular ROS and telomere DNA damage. Mice between 4 weeks and 20 months of age were studied. |
Nfkb1, Rela [156] | Nfkb1-KO and Rela-KO mice. | Hepatic steatosis Hepatic steatosis was decreased in Nfkb1-KO mice; also, they showed reduction of both lipogenic genes (scd1, Fas and Cd36) and lipogenic proteins (SREBP1c and SCD1). Implication of a signaling pathway of Nfkb1-/HDAC1/SCREBP1c in hepatocytes in the control of hepatic steatosis. Inactivation of NF-κB p65 did not alter the hepatic steatosis. Mice between 8 and 28 weeks of age were studied. |
Cyld [66] | K5-CyldC/S mice. Expression of a mutant Cyld gene carrying a point mutation C/S that acts as a dominant negative in keratin 5 expressing cells. | Chronic hepatitis K5-CyldC/S exhibit signs of premature aged liver, generalized inflammation, with multifocal chronic hepatitis; develop spontaneous HCC and hepatocellular adenomas. Chronic NF-κBp65 activation. Tumors likely develop as a consequence both of the premature aging and the systemic inflammation of the transgenic mice. Young (1- to 9-month-old) and old (20- to 24-month-old) mice were analyzed. |
Nik, Ikkα [165] | Hepatocyte-specific Nik (NikDhep) and Ikkα (ikkαDhep) knockout mice. | Progression of liver disease Pathological activation of hepatic NIK or IKKα likely blocks hepatocyte replication, contributing to liver disease progression in PHx mouse models. The NIK/IKKα pathway suppresses reparative hepatocyte proliferation at least in part by inhibiting the JAK2/STAT3 pathway. Mice 8 and 18 weeks of age were studied. |
NF-κB Gene | Genetic Modification | Involvement of NF-κB in the Ageing of Progenitor Cells. Proposed Mechanisms |
---|---|---|
Muscle-derived stem/progenitor cells | ||
Rela [172] | Rela haploinsufficient (aged p65+/−) mice. | Aged p65+/− MDSPCs retained myogenic potential in vitro and had a higher resistance to oxidative stress-induced cell death. Genetic inhibition of NF-κB activation increases myogenic differentiation and improves resistance to oxidative stress. 14-day-old and 24-month-old mice were studied. |
Ikkβ, nemo [173] | IkkβCAMuSC mice express a constitutively active (CA) form of the Ikkβ gene in MuSCs. NemoKOMuSC: Mice deficient in Nemo subunit in MuSCs. Mdx mice: Dystrophin mutant mouse. | Persistent activation of NF-κB in IkkβCAMuSC mice leads to telomere shortening and impairment of regeneration; in Mdx/IkkβCAMuSC mice it exacerbates the progression of dystrophy. The mechanism involves Ku80 dysregulation (a protein that binds to telomeres, aid the localization of other shelterins, and regulate telomere length) and increased DNA damage, specifically on telomeres, resulting in MuSC loss and, subsequently, skeletal muscle regenerative failure. Mice between 2- and 12 months of age were analyzed. |
Osteoprogenitors | ||
Ikkβ [107] | iNf-κb/OP mice: express a constitutively active mutant IKKβ in osteoprogenitor (OP)-lineage cells, upon Dox withdrawal in skeletally mature mice. | iNf-κb/OP mice showed reduced bone mineral density in the femur and tibia, and increased bone marrow fat, resembling human osteoporosis. Decreased expression levels of osteogenic marker (Runx2 and osteocalcin) and increased adipogenic marker (PPAR-γ and C/EBP) in mesenchymal stem cells (MSCs) of iNf-κb/OP mice. Mice of 12 and 36 weeks of age were studied. |
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
García-García, V.A.; Alameda, J.P.; Page, A.; Casanova, M.L. Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells 2021, 10, 1906. https://doi.org/10.3390/cells10081906
García-García VA, Alameda JP, Page A, Casanova ML. Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells. 2021; 10(8):1906. https://doi.org/10.3390/cells10081906
Chicago/Turabian StyleGarcía-García, Verónica A., Josefa P. Alameda, Angustias Page, and María Llanos Casanova. 2021. "Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models" Cells 10, no. 8: 1906. https://doi.org/10.3390/cells10081906
APA StyleGarcía-García, V. A., Alameda, J. P., Page, A., & Casanova, M. L. (2021). Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells, 10(8), 1906. https://doi.org/10.3390/cells10081906