301
Views
3
CrossRef citations to date
0
Altmetric
Review

The Relationship Between Alzheimer’s Disease and Skin Diseases: A Review

, , & ORCID Icon
Pages 1551-1560 | Published online: 24 Oct 2021

References

  • Zhang H, Chen K, Wang N, et al. Analysis of brain donors’ demographic and medical characteristics to facilitate the construction of a human brain bank in China. J Alzheimers Dis. 2018;66(3):1245–1254. doi:10.3233/JAD-180779
  • Goodman RA, Lochner KA, Thambisetty M, Wingo TS, Posner SF, Ling SM. Prevalence of dementia subtypes in United States Medicare fee-for-service beneficiaries, 2011–2013. Alzheimers Dement. 2017;13(1):28–37. doi:10.1016/j.jalz.2016.04.002
  • Scheltens P, De Strooper B, Kivipelto M, et al. Alzheimer’s disease. Lancet. 2021;397(10284):1577–1590. doi:10.1016/S0140-6736(20)32205-4
  • Clos AL, Kayed R, Lasagna-Reeves CA. Association of skin with the pathogenesis and treatment of neurodegenerative amyloidosis. Front Neurol. 2012;3:5. doi:10.3389/fneur.2012.00005
  • Akerman SC, Hossain S, Shobo A, et al. Neurodegenerative disease-related proteins within the epidermal layer of the human skin. J Alzheimers Dis. 2019;69(2):463–478. doi:10.3233/JAD-181191
  • Försti AK, Jokelainen J, Ansakorpi H, et al. Psychiatric and neurological disorders are associated with bullous pemphigoid - a nationwide Finnish Care Register study. Sci Rep. 2016;6:37125. doi:10.1038/srep37125
  • White RS, Lipton RB, Hall CB, Steinerman JR. Nonmelanoma skin cancer is associated with reduced Alzheimer disease risk. Neurology. 2013;80(21):1966–1972. doi:10.1212/WNL.0b013e3182941990
  • Kluger N, Pankakoski A, Panelius J. Depression and anxiety in patients with bullous pemphigoid: impact and management challenges. Clin Cosmet Investig Dermatol. 2020;13:73–76. doi:10.2147/CCID.S212984
  • Delli FS, Sotiriou E, Lazaridou E, et al. Total IgE, eosinophils, and interleukins 16, 17A, and 23 correlations in severe bullous pemphigoid and treatment implications. Dermatol Ther. 2020;33(6):e13958. doi:10.1111/dth.13958
  • Azimi SZ, Firooz A, Murrell DF, Daneshpazhooh M. Treatment concerns for bullous pemphigoid in the COVID-19 pandemic era. Dermatol Ther. 2020;33(6):e13956. doi:10.1111/dth.13956
  • Wang YN, Hammers CM, Mao X, Jin HZ, Yuan J, Li L. Analysis of the autoimmune response against BP180 in patients with Alzheimer’s disease. Ann Transl Med. 2021;9(2):107. doi:10.21037/atm-20-5343
  • Zhao W, Wang Y, Mao X, et al. Detection of underlying dementia in bullous pemphigoid patients using cognitive evaluation tests: a multicenter case-control study. Ann Transl Med. 2020;8(21):1397. doi:10.21037/atm-20-1319
  • Schmidt E, Zillikens D. Pemphigoid diseases. Lancet. 2013;381(9863):320–332. doi:10.1016/S0140-6736(12)61140-4
  • Langan SM, Groves RW, West J. The relationship between neurological disease and bullous pemphigoid: a population-based case-control study. J Invest Dermatol. 2011;131(3):631–636. doi:10.1038/jid.2010.357
  • Lai YC, Yew YW, Lambert WC. Bullous pemphigoid and its association with neurological diseases: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2016;30(12):2007–2015. doi:10.1111/jdv.13660
  • Taghipour K, Chi CC, Vincent A, Groves RW, Venning V, Wojnarowska F. The association of bullous pemphigoid with cerebrovascular disease and dementia: a case-control study. Arch Dermatol. 2010;146(11):1251–1254. doi:10.1001/archdermatol.2010.322
  • Messingham KN, Miller AD, Narayanan NS, Connell SJ, Fairley JA. Demographics and autoantibody profiles of pemphigoid patients with underlying neurologic diseases. J Invest Dermatol. 2019;139(9):1860–1866. doi:10.1016/j.jid.2019.01.034
  • Bastuji-Garin S, Joly P, Lemordant P, et al. Risk factors for bullous pemphigoid in the elderly: a prospective case-control study. J Invest Dermatol. 2011;131(3):637–643. doi:10.1038/jid.2010.301
  • Chen YJ, Wu CY, Lin MW, et al. Comorbidity profiles among patients with bullous pemphigoid: a nationwide population-based study. Br J Dermatol. 2011;165(3):593–599. doi:10.1111/j.1365-2133.2011.10386.x
  • Brick KE, Weaver CH, Savica R, et al. A population-based study of the association between bullous pemphigoid and neurologic disorders. J Am Acad Dermatol. 2014;71(6):1191–1197. doi:10.1016/j.jaad.2014.07.052
  • Kibsgaard L, Rasmussen M, Lamberg A, Deleuran M, Olesen AB, Vestergaard C. Increased frequency of multiple sclerosis among patients with bullous pemphigoid: a population-based cohort study on comorbidities anchored around the diagnosis of bullous pemphigoid. Br J Dermatol. 2017;176(6):1486–1491. doi:10.1111/bjd.15405
  • Papakonstantinou E, Limberg MM, Gehring M, et al. Neurological disorders are associated with bullous pemphigoid. J Eur Acad Dermatol Venereol. 2019;33(5):925–929. doi:10.1111/jdv.15444
  • Seppänen A, Suuronen T, Hofmann SC, Majamaa K, Alafuzoff I. Distribution of collagen XVII in the human brain. Brain Res. 2007;1158:50–56. doi:10.1016/j.brainres.2007.04.073
  • Seppänen A, Autio-Harmainen H, Alafuzoff I, et al. Collagen XVII is expressed in human CNS neurons. Matrix Biol. 2006;25(3):185–188. doi:10.1016/j.matbio.2005.11.004
  • Kridin K, Hübner F, Recke A, Linder R, Schmidt E. The burden of neurological comorbidities in six autoimmune bullous diseases: a population-based study. J Eur Acad Dermatol Venereol. 2021. doi:10.1111/jdv.17465
  • Seppänen A. Collagen XVII: a shared antigen in neurodermatological interactions? Clin Dev Immunol. 2013;2013:240570. doi:10.1155/2013/240570
  • Chen J, Li L, Chen J, et al. Sera of elderly bullous pemphigoid patients with associated neurological diseases recognize bullous pemphigoid antigens in the human brain. Gerontology. 2011;57(3):211–216. doi:10.1159/000315393
  • Kokkonen N, Herukka SK, Huilaja L, et al. Increased levels of the bullous pemphigoid BP180 autoantibody are associated with more severe dementia in Alzheimer’s disease. J Invest Dermatol. 2017;137(1):71–76. doi:10.1016/j.jid.2016.09.010
  • Messingham KAN, Aust S, Helfenberger J, et al. Autoantibodies to collagen XVII are present in Parkinson’s disease and localize to tyrosine-hydroxylase positive neurons. J Invest Dermatol. 2016;136(3):721–723. doi:10.1016/j.jid.2015.12.005
  • Recke A, Oei A, Hübner F, et al. Parkinson disease and multiple sclerosis are not associated with autoantibodies against structural proteins of the dermal-epidermal junction. Br J Dermatol. 2016;175(2):407–409. doi:10.1111/bjd.14538
  • Barrick BJ, Ida CM, Laniosz V, et al. Bullous pemphigoid, neurodegenerative disease, and hippocampal BP180 expression: a retrospective postmortem neuropathologic study. J Invest Dermatol. 2016;136(10):2090–2092. doi:10.1016/j.jid.2016.06.015
  • Künzli K, Favre B, Chofflon M, Borradori L. One gene but different proteins and diseases: the complexity of dystonin and bullous pemphigoid antigen 1. Exp Dermatol. 2016;25(1):10–16. doi:10.1111/exd.12877
  • Tuusa J, Lindgren O, Tertsunen HM, et al. BP180 autoantibodies target different epitopes in multiple sclerosis or Alzheimer’s disease than in bullous pemphigoid. J Invest Dermatol. 2019;139(2):293–299. doi:10.1016/j.jid.2018.09.010
  • Amber KT, Zikry J, Hertl M. A multi-hit hypothesis of bullous pemphigoid and associated neurological disease: is HLA-DQB1*03:01, a potential link between immune privileged antigen exposure and epitope spreading? Hla. 2017;89(3):127–134. doi:10.1111/tan.12960
  • Koumaki D, Efthymiou O, Bozi E, Katoulis AC. Perspectives on perceived stigma and self-stigma in patients with hidradenitis suppurativa. Clin Cosmet Investig Dermatol. 2019;12:785–790. doi:10.2147/CCID.S180036
  • Fernandez JM, Marr KD, Hendricks AJ, et al. Alleviating and exacerbating foods in hidradenitis suppurativa. Dermatol Ther. 2020;33(6):e14246. doi:10.1111/dth.14246
  • Misitzis A, Goldust M, Jafferany M, Lotti T. Psychiatric comorbidities in patients with hidradenitis suppurativa. Dermatol Ther. 2020;33(4):e13541. doi:10.1111/dth.13541
  • Shah M, Sachdeva M, Melendez-Gonzalez M, Piguet V, Sayed C. Hidradenitis suppurativa and chromosomal abnormalities: a case report and systematic review of the literature. Int J Dermatol. 2021;60(3):261–271. doi:10.1111/ijd.15111
  • Sabat R, Jemec GBE, Matusiak Ł, Kimball AB, Prens E, Wolk K. Hidradenitis suppurativa. Nat Rev Dis Primers. 2020;6(1):18. doi:10.1038/s41572-020-0149-1
  • Nguyen TV, Damiani G, Orenstein LAV, Hamzavi I, Jemec GB. Hidradenitis suppurativa: an update on epidemiology, phenotypes, diagnosis, pathogenesis, comorbidities and quality of life. J Eur Acad Dermatol Venereol. 2021;35(1):50–61. doi:10.1111/jdv.16677
  • De Strooper B, Iwatsubo T, Wolfe MS. Presenilins and γ-secretase: structure, function, and role in Alzheimer Disease. Cold Spring Harb Perspect Med. 2012;2(1):a006304. doi:10.1101/cshperspect.a006304
  • Ingram JR. The genetics of hidradenitis suppurativa. Dermatol Clin. 2016;34(1):23–28. doi:10.1016/j.det.2015.07.002
  • Panza F, Frisardi V, Imbimbo BP, et al. REVIEW: γ-Secretase inhibitors for the treatment of Alzheimer’s disease: the current state. CNS Neurosci Ther. 2010;16(5):272–284. doi:10.1111/j.1755-5949.2010.00164.x
  • Xia X, Qian S, Soriano S, et al. Loss of presenilin 1 is associated with enhanced beta-catenin signaling and skin tumorigenesis. Proc Natl Acad Sci U S A. 2001;98(19):10863–10868. doi:10.1073/pnas.191284198
  • Pan Y, Lin MH, Tian X, et al. γ-Secretase functions through notch signaling to maintain skin appendages but is not required for their patterning or initial morphogenesis. Dev Cell. 2004;7(5):731–743. doi:10.1016/j.devcel.2004.09.014
  • Li T, Wen H, Brayton C, et al. Epidermal growth factor receptor and notch pathways participate in the tumor suppressor function of γ-secretase. J Biol Chem. 2007;282(44):32264–32273. doi:10.1074/jbc.M703649200
  • Girouard SD, Falk RH, Rennke HG, Merola JF. Hidradenitis suppurativa resulting in systemic amyloid A amyloidosis: a case report and review of the literature. Dermatol Online J. 2012;18(1):2. doi:10.5070/D30P46V20Q
  • Canoui-Poitrine F, Le Thuaut A, Revuz JE, et al. Identification of three hidradenitis suppurativa phenotypes: latent class analysis of a cross-sectional study. J Invest Dermatol. 2013;133(6):1506–1511. doi:10.1038/jid.2012.472
  • Ingram JR, Piguet V. Phenotypic heterogeneity in hidradenitis suppurativa (acne inversa): classification is an essential step toward personalized therapy. J Invest Dermatol. 2013;133(6):1453–1456. doi:10.1038/jid.2012.476
  • Pink AE, Simpson MA, Desai N, et al. Mutations in the γ-secretase genes NCSTN, PSENEN, and PSEN1 underlie rare forms of hidradenitis suppurativa (acne inversa). J Invest Dermatol. 2012;132(10):2459–2461. doi:10.1038/jid.2012.162
  • Ingram JR, Wood M, John B, Butler R, Anstey AV. Absence of pathogenic γ-secretase mutations in a South Wales cohort of familial and sporadic hidradenitis suppurativa (acne inversa). Br J Dermatol. 2013;168(4):874–876. doi:10.1111/bjd.12048
  • Nomura Y, Nomura T, Suzuki S, et al. A novel NCSTN mutation alone may be insufficient for the development of familial hidradenitis suppurativa. J Dermatol Sci. 2014;74(2):180–182. doi:10.1016/j.jdermsci.2014.01.013
  • Garg A, Strunk A. Risk of Alzheimer’s disease is not increased among patients with hidradenitis suppurativa: a retrospective population-based cohort analysis. J Am Acad Dermatol. 2017;77(1):176–177. doi:10.1016/j.jaad.2017.02.055
  • Theut Riis P, Egeberg A, Gislason GH, Jemec GB. Patients with hidradenitis suppurativa have no increased risk of Alzheimer disease. Br J Dermatol. 2017;177(1):273–275. doi:10.1111/bjd.15064
  • Frew JW, Vekic DA, Woods J, Cains GD. A systematic review and critical evaluation of reported pathogenic sequence variants in hidradenitis suppurativa. Br J Dermatol. 2017;177(4):987–998. doi:10.1111/bjd.15441
  • Frew JW, Navrazhina K. In silico analysis of gamma-secretase-complex mutations in hidradenitis suppurativa demonstrates disease-specific substrate recognition and cleavage alterations. Front Med. 2019;6:206. doi:10.3389/fmed.2019.00206
  • Li A, Peng Y, Taiclet LM, Tanzi RE. Analysis of hidradenitis suppurativa-linked mutations in four genes and the effects of PSEN1-P242LfsX11 on cytokine and chemokine expression in macrophages. Hum Mol Genet. 2019;28(7):1173–1182. doi:10.1093/hmg/ddy414
  • Esme P, Esme M, Caliskan E. Increased prevalence of family history of Alzheimer’s disease in hidradenitis suppurativa: cross-sectional analysis of 192 HS patients. Dermatol Ther. 2020;33(6):e14219. doi:10.1111/dth.14219
  • Nada EA, Muhammad EMS, Ahmed SFM, Tamam AMS, Abdelhamed A. Assessment of the effect of metabolic syndrome on the autophagy marker LC3 in psoriasis vulgaris patients: a cross-sectional study. Clin Cosmet Investig Dermatol. 2020;13:1005–1013. doi:10.2147/CCID.S284300
  • Carrascosa JM, Theng C, Thaçi D. Spotlight on topical long-term management of plaque psoriasis. Clin Cosmet Investig Dermatol. 2020;13:495–498. doi:10.2147/CCID.S254114
  • Fu Y, Lee CH, Chi CC. Association of psoriasis with inflammatory bowel disease: a systematic review and meta-analysis. JAMA Dermatol. 2018;154(12):1417–1423. doi:10.1001/jamadermatol.2018.3631
  • Soliman M. Patient-reported disease severity and quality of life among Arabic psoriatic patients: a cross-sectional survey. Clin Cosmet Investig Dermatol. 2020;13:601–609. doi:10.2147/CCID.S269909
  • Ezzedine K, Fougerousse AC, Aubert R, et al. Individual burden of psoriasis (I-BOP): building and validation of a new scoring tool for patients with psoriasis. Clin Cosmet Investig Dermatol. 2020;13:325–332. doi:10.2147/CCID.S249776
  • Orrell KA, Vakharia PP, Hagstrom EL, Brieva J, West DP, Nardone B. Prevalence of chronic hepatitis B and C in psoriasis patients: a cross-sectional study in a large US population. J Am Acad Dermatol. 2017;77(3):572–573. doi:10.1016/j.jaad.2017.05.020
  • Leisner MZ, Riis JL, Schwartz S, Iversen L, Østergaard SD, Olsen MS. Psoriasis and risk of mental disorders in Denmark. JAMA Dermatol. 2019;155(6):745–747. doi:10.1001/jamadermatol.2019.0039
  • Kim M, Park HE, Lee SH, Han K, Lee JH. Increased risk of Alzheimer’s disease in patients with psoriasis: a nationwide population-based cohort study. Sci Rep. 2020;10(1):6454. doi:10.1038/s41598-020-63550-2
  • Huang KL, Yeh CC, Wu SI, et al. Risk of dementia among individuals with psoriasis: a nationwide population-based cohort study in Taiwan. J Clin Psychiatry. 2019;80(3):18m12462. doi:10.4088/JCP.18m12462
  • Lin CC, Lin HC, Chiu HW. Association between psoriasis and dementia: a population-based case-control study. Am J Clin Dermatol. 2019;20(3):457–463. doi:10.1007/s40257-018-00420-8
  • Pezzolo E, Mutlu U, Vernooij MW, et al. Psoriasis is not associated with cognition, brain imaging markers and risk of dementia: the Rotterdam Study. J Am Acad Dermatol. 2018. doi:10.1016/j.jaad.2018.07.046
  • Liu L, Chen ST, Li HJ, et al. Association between psoriasis and dementia: current evidence. Front Aging Neurosci. 2020;12:570992. doi:10.3389/fnagi.2020.570992
  • Abuabara K, Azfar RS, Shin DB, Neimann AL, Troxel AB, Gelfand JM. Cause-specific mortality in patients with severe psoriasis: a population-based cohort study in the U.K. Br J Dermatol. 2010;163(3):586–592. doi:10.1111/j.1365-2133.2010.09941.x
  • Gisondi P, Sala F, Alessandrini F, et al. Mild cognitive impairment in patients with moderate to severe chronic plaque psoriasis. Dermatology. 2014;228(1):78–85. doi:10.1159/000357220
  • Colgecen E, Celikbilek A, Keskin DT. Cognitive impairment in patients with psoriasis: a cross-sectional study using the Montreal cognitive assessment. Am J Clin Dermatol. 2016;17(4):413–419. doi:10.1007/s40257-016-0187-3
  • Decourt B, Lahiri DK, Sabbagh MN. Targeting tumor necrosis factor alpha for Alzheimer’s disease. Curr Alzheimer Res. 2017;14(4):412–425. doi:10.2174/1567205013666160930110551
  • Zhou M, Xu R, Kaelber DC, Gurney ME. Tumor necrosis factor (TNF) blocking agents are associated with lower risk for Alzheimer’s disease in patients with rheumatoid arthritis and psoriasis. PLoS One. 2020;15(3):e0229819. doi:10.1371/journal.pone.0229819
  • Rich P, Bourcier M, Sofen H, et al. Ustekinumab improves nail disease in patients with moderate-to-severe psoriasis: results from PHOENIX 1. Br J Dermatol. 2014;170(2):398–407. doi:10.1111/bjd.12632
  • Mohammadi Shahrokhi V, Ravari A, Mirzaei T, Zare-Bidaki M, Asadikaram G, Arababadi MK. IL-17A and IL-23: plausible risk factors to induce age-associated inflammation in Alzheimer’s disease. Immunol Invest. 2018;47(8):812–822. doi:10.1080/08820139.2018.1504300
  • Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16(6):358–372. doi:10.1038/nrn3880
  • Vom Berg J, Prokop S, Miller KR, et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat Med. 2012;18(12):1812–1819. doi:10.1038/nm.2965
  • Belloy ME, Napolioni V, Greicius MD. A quarter century of APOE and Alzheimer’s disease: progress to date and the path forward. Neuron. 2019;101(5):820–838. doi:10.1016/j.neuron.2019.01.056
  • Al Harthi F, Huraib GB, Zouman A, Arfin M, Tariq M, Al-Asmari A. Apolipoprotein E gene polymorphism and serum lipid profile in Saudi patients with psoriasis. Dis Markers. 2014;2014:239645. doi:10.1155/2014/239645
  • Campalani E, Allen MH, Fairhurst D, et al. Apolipoprotein E gene polymorphisms are associated with psoriasis but do not determine disease response to acitretin. Br J Dermatol. 2006;154(2):345–352. doi:10.1111/j.1365-2133.2005.06950.x
  • Han Y, Liu T, Lu L. Apolipoprotein E gene polymorphism in psoriasis: a meta-analysis. Arch Med Res. 2013;44(1):46–53. doi:10.1016/j.arcmed.2012.10.009
  • Coto-Segura P, Coto E, Alvarez V, et al. Apolipoprotein ε4 allele is associated with psoriasis severity. Arch Dermatol Res. 2010;302(2):145–149. doi:10.1007/s00403-009-1002-2
  • Yokoyama JS, Wang Y, Schork AJ, et al. Association between genetic traits for immune-mediated diseases and Alzheimer disease. JAMA Neurol. 2016;73(6):691–697. doi:10.1001/jamaneurol.2016.0150
  • Ospina-Romero M, Glymour MM, Hayes-Larson E, et al. Association between Alzheimer disease and cancer with evaluation of study biases: a systematic review and meta-analysis. JAMA Network Open. 2020;3(11):e2025515. doi:10.1001/jamanetworkopen.2020.25515
  • Roe CM, Fitzpatrick AL, Xiong C, et al. Cancer linked to Alzheimer disease but not vascular dementia. Neurology. 2010;74(2):106–112. doi:10.1212/WNL.0b013e3181c91873
  • Leiter U, Keim U, Garbe C. Epidemiology of skin cancer: update 2019. Adv Exp Med Biol. 2020;1268:123–139.
  • Schmidt SA, Ording AG, Horváth-Puhó E, Sørensen HT, Henderson VW. Non-melanoma skin cancer and risk of Alzheimer’s disease and all-cause dementia. PLoS One. 2017;12(2):e0171527. doi:10.1371/journal.pone.0171527
  • Ibler E, Tran G, Orrell KA, et al. Inverse association for diagnosis of Alzheimer’s disease subsequent to both melanoma and non-melanoma skin cancers in a large, urban, single-centre, Midwestern US patient population. J Eur Acad Dermatol Venereol. 2018;32(11):1893–1896. doi:10.1111/jdv.14952
  • Houck AL, Seddighi S, Driver JA. At the crossroads between neurodegeneration and cancer: a review of overlapping biology and its implications. Curr Aging Sci. 2018;11(2):77–89. doi:10.2174/1874609811666180223154436
  • Nakanishi A, Minami A, Kitagishi Y, Ogura Y, Matsuda S. BRCA1 and p53 tumor suppressor molecules in Alzheimer’s disease. Int J Mol Sci. 2015;16(2):2879–2892. doi:10.3390/ijms16022879
  • Ma SL, Tang NL, Tam CW, et al. A PIN1 polymorphism that prevents its suppression by AP4 associates with delayed onset of Alzheimer’s disease. Neurobiol Aging. 2012;33(4):804–813. doi:10.1016/j.neurobiolaging.2010.05.018
  • Segat L, Pontillo A, Annoni G, et al. PIN1 promoter polymorphisms are associated with Alzheimer’s disease. Neurobiol Aging. 2007;28(1):69–74. doi:10.1016/j.neurobiolaging.2005.11.009
  • Kumar S, Reddy PH. A new discovery of MicroRNA-455-3p in Alzheimer’s disease. J Alzheimers Dis. 2019;72(s1):S117–s130. doi:10.3233/JAD-190583
  • Galvão F, Grokoski KC, da Silva BB, Lamers ML, Siqueira IR. The amyloid precursor protein (APP) processing as a biological link between Alzheimer’s disease and cancer. Ageing Res Rev. 2019;49:83–91. doi:10.1016/j.arr.2018.11.007
  • Botelho MG, Wang X, Arndt-Jovin DJ, Becker D, Jovin TM. Induction of terminal differentiation in melanoma cells on downregulation of β-amyloid precursor protein. J Invest Dermatol. 2010;130(5):1400–1410. doi:10.1038/jid.2009.296
  • Matafora V, Farris F, Restuccia U, et al. Amyloid aggregates accumulate in melanoma metastasis modulating YAP activity. EMBO Rep. 2020;21(9):e50446. doi:10.15252/embr.202050446
  • Cummings J. What can be inferred from the interruption of the semagacestat trial for treatment of Alzheimer’s disease? Biol Psychiatry. 2010;68(10):876–878. doi:10.1016/j.biopsych.2010.09.020
  • Li T, Wen H, Brayton C, et al. Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. J Neurosci. 2007;27(40):10849–10859. doi:10.1523/JNEUROSCI.2152-07.2007
  • Ostendorf BN, Bilanovic J, Adaku N, et al. Common germline variants of the human APOE gene modulate melanoma progression and survival. Nat Med. 2020;26(7):1048–1053. doi:10.1038/s41591-020-0879-3
  • Lövdén M, Xu W, Wang HX. Lifestyle change and the prevention of cognitive decline and dementia: what is the evidence? Curr Opin Psychiatry. 2013;26(3):239–243. doi:10.1097/YCO.0b013e32835f4135
  • D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T. UV radiation and the skin. Int J Mol Sci. 2013;14(6):12222–12248. doi:10.3390/ijms140612222
  • Steding-Jessen M, Birch-Johansen F, Jensen A, Schüz J, Kjær SK, Dalton SO. Socioeconomic status and non-melanoma skin cancer: a nationwide cohort study of incidence and survival in Denmark. Cancer Epidemiol. 2010;34(6):689–695. doi:10.1016/j.canep.2010.06.011
  • Tillement JP, Lecanu L, Papadopoulos V. Amyloidosis and neurodegenerative diseases: current treatments and new pharmacological options. Pharmacology. 2010;85(1):1–17. doi:10.1159/000259044
  • Schreml S, Kaiser E, Landthaler M, Szeimies RM, Babilas P. Amyloid in skin and brain: what’s the link? Exp Dermatol. 2010;19(11):953–957. doi:10.1111/j.1600-0625.2010.01166.x
  • Awal G, Kaur S. Association of cutaneous amyloidosis with neurodegenerative amyloidosis: correlation or coincidence? J Clin Aesthet Dermatol. 2018;11(4):25–27.
  • Chen G, Chen ZM, Fan XY, et al. Gut-brain-skin axis in psoriasis: a review. Dermatol Ther. 2021;11(1):25–38. doi:10.1007/s13555-020-00466-9
  • Neau JP, Godeneche G, Mathis S, Guillet G. Neurodermatology. Handb Clin Neurol. 2014;121:1561–1594.