메뉴 건너뛰기




Volumn 145, Issue 4, 2016, Pages 401-417

Molecular insights into the premature aging disease progeria

Author keywords

Adult stem cells; Chromatin; Lamins; Nucleoplasmic lamins; Premature aging; Progeria; Senescence; Signaling

Indexed keywords

LAMIN; LAMIN A; LMNA PROTEIN, HUMAN;

EID: 84957573722     PISSN: 09486143     EISSN: 1432119X     Source Type: Journal    
DOI: 10.1007/s00418-016-1411-1     Document Type: Review
Times cited : (85)

References (181)
  • 1
    • 84897036591 scopus 로고    scopus 로고
    • Mechanisms and dynamics of nuclear lamina–genome interactions
    • COI: 1:CAS:528:DC%2BC2cXhtVSlsbzE, PID: 24694724
    • Amendola M, van Steensel B (2014) Mechanisms and dynamics of nuclear lamina–genome interactions. Curr Opin Cell Biol 28:61–68. doi:10.1016/j.ceb.2014.03.003
    • (2014) Curr Opin Cell Biol , vol.28 , pp. 61-68
    • Amendola, M.1    van Steensel, B.2
  • 2
    • 76149084859 scopus 로고    scopus 로고
    • Role of A-type lamins in signaling, transcription, and chromatin organization
    • COI: 1:CAS:528:DC%2BC3cXhs1KlsA%3D%3D, PID: 20038676
    • Andres V, Gonzalez JM (2009) Role of A-type lamins in signaling, transcription, and chromatin organization. J Cell Biol 187(7):945–957. doi:10.1083/jcb.200904124
    • (2009) J Cell Biol , vol.187 , Issue.7 , pp. 945-957
    • Andres, V.1    Gonzalez, J.M.2
  • 4
    • 77952009403 scopus 로고    scopus 로고
    • Quantification of hemodynamic wall shear stress in patients with bicuspid aortic valve using phase-contrast MRI
    • PID: 19953319
    • Barker AJ, Lanning C, Shandas R (2010) Quantification of hemodynamic wall shear stress in patients with bicuspid aortic valve using phase-contrast MRI. Ann Biomed Eng 38(3):788–800. doi:10.1007/s10439-009-9854-3
    • (2010) Ann Biomed Eng , vol.38 , Issue.3 , pp. 788-800
    • Barker, A.J.1    Lanning, C.2    Shandas, R.3
  • 6
    • 77955385716 scopus 로고    scopus 로고
    • Role of progerin-induced telomere dysfunction in HGPS premature cellular senescence
    • COI: 1:CAS:528:DC%2BC3cXhtF2mtL7J, PID: 20605919
    • Benson EK, Lee SW, Aaronson SA (2010) Role of progerin-induced telomere dysfunction in HGPS premature cellular senescence. J Cell Sci 123(15):2605–2612. doi:10.1242/jcs.067306
    • (2010) J Cell Sci , vol.123 , Issue.15 , pp. 2605-2612
    • Benson, E.K.1    Lee, S.W.2    Aaronson, S.A.3
  • 8
    • 22944488871 scopus 로고    scopus 로고
    • Telomeres and human disease: ageing, cancer and beyond
    • COI: 1:CAS:528:DC%2BD2MXntFeqtbw%3D, PID: 16136653
    • Blasco MA (2005) Telomeres and human disease: ageing, cancer and beyond. Nat Rev Genet 6(8):611–622. doi:10.1038/nrg1656
    • (2005) Nat Rev Genet , vol.6 , Issue.8 , pp. 611-622
    • Blasco, M.A.1
  • 9
    • 81855217987 scopus 로고    scopus 로고
    • Evolvement of LEM proteins as chromatin tethers at the nuclear periphery
    • COI: 1:CAS:528:DC%2BC3MXhsFaiur3N, PID: 22103517
    • Brachner A, Foisner R (2011) Evolvement of LEM proteins as chromatin tethers at the nuclear periphery. Biochem Soc Trans 39(6):1735–1741. doi:10.1042/BST20110724
    • (2011) Biochem Soc Trans , vol.39 , Issue.6 , pp. 1735-1741
    • Brachner, A.1    Foisner, R.2
  • 10
    • 84955329648 scopus 로고    scopus 로고
    • Hutchinson–Gilford progeria syndrome as a model for vascular aging
    • PID: 26330290
    • Brassard JA, Fekete N, Garnier A, Hoesli CA (2015) Hutchinson–Gilford progeria syndrome as a model for vascular aging. Biogerontology. doi:10.1007/s10522-015-9602-z
    • (2015) Biogerontology
    • Brassard, J.A.1    Fekete, N.2    Garnier, A.3    Hoesli, C.A.4
  • 11
    • 84874591240 scopus 로고    scopus 로고
    • The role of mitochondria in aging
    • COI: 1:CAS:528:DC%2BC3sXjvVyhsL8%3D, PID: 23454757
    • Bratic A, Larsson NG (2013) The role of mitochondria in aging. J Clin Invest 123(3):951–957. doi:10.1172/JCI64125
    • (2013) J Clin Invest , vol.123 , Issue.3 , pp. 951-957
    • Bratic, A.1    Larsson, N.G.2
  • 12
    • 2342644879 scopus 로고    scopus 로고
    • Aging of Hutchinson–Gilford progeria syndrome fibroblasts is characterised by hyperproliferation and increased apoptosis
    • COI: 1:CAS:528:DC%2BD2cXjvVahtbc%3D, PID: 15130666
    • Bridger JM, Kill IR (2004) Aging of Hutchinson–Gilford progeria syndrome fibroblasts is characterised by hyperproliferation and increased apoptosis. Exp Gerontol 39(5):717–724
    • (2004) Exp Gerontol , vol.39 , Issue.5 , pp. 717-724
    • Bridger, J.M.1    Kill, I.R.2
  • 13
    • 33745915850 scopus 로고    scopus 로고
    • Nuclear lamins: laminopathies and their role in premature ageing
    • COI: 1:CAS:528:DC%2BD28XnsFGgtbg%3D, PID: 16816143
    • Broers JL, Ramaekers FC, Bonne G, Yaou RB, Hutchison CJ (2006) Nuclear lamins: laminopathies and their role in premature ageing. Physiol Rev 86(3):967–1008. doi:10.1152/physrev.00047.2005
    • (2006) Physiol Rev , vol.86 , Issue.3 , pp. 967-1008
    • Broers, J.L.1    Ramaekers, F.C.2    Bonne, G.3    Yaou, R.B.4    Hutchison, C.J.5
  • 15
    • 84930680704 scopus 로고    scopus 로고
    • Medicine. Progeria accelerates adult stem cell aging
    • COI: 1:CAS:528:DC%2BC2MXhtVGntLbJ, PID: 26045423
    • Brunauer R, Kennedy BK (2015) Medicine. Progeria accelerates adult stem cell aging. Science 348(6239):1093–1094. doi:10.1126/science.aac4214
    • (2015) Science , vol.348 , Issue.6239 , pp. 1093-1094
    • Brunauer, R.1    Kennedy, B.K.2
  • 16
    • 77954950116 scopus 로고    scopus 로고
    • Progeria syndromes and ageing: what is the connection?
    • COI: 1:CAS:528:DC%2BC3cXpt1art7s%3D, PID: 20651707
    • Burtner CR, Kennedy BK (2010) Progeria syndromes and ageing: what is the connection? Nat Rev Mol Cell Biol 11(8):567–578. doi:10.1038/nrm2944
    • (2010) Nat Rev Mol Cell Biol , vol.11 , Issue.8 , pp. 567-578
    • Burtner, C.R.1    Kennedy, B.K.2
  • 19
    • 80855128461 scopus 로고    scopus 로고
    • Cellular senescence: a link between cancer and age-related degenerative disease?
    • COI: 1:CAS:528:DC%2BC3MXhsFOhtb7F, PID: 21925603
    • Campisi J, Andersen JK, Kapahi P, Melov S (2011) Cellular senescence: a link between cancer and age-related degenerative disease? Semin Cancer Biol 21(6):354–359. doi:10.1016/j.semcancer.2011.09.001
    • (2011) Semin Cancer Biol , vol.21 , Issue.6 , pp. 354-359
    • Campisi, J.1    Andersen, J.K.2    Kapahi, P.3    Melov, S.4
  • 20
    • 79959928025 scopus 로고    scopus 로고
    • Progerin and telomere dysfunction collaborate to trigger cellular senescence in normal human fibroblasts
    • COI: 1:CAS:528:DC%2BC3MXovVKgu7k%3D, PID: 21670498
    • Cao K, Blair CD, Faddah DA, Kieckhaefer JE, Olive M, Erdos MR, Nabel EG, Collins FS (2011a) Progerin and telomere dysfunction collaborate to trigger cellular senescence in normal human fibroblasts. J Clin Invest 121(7):2833–2844. doi:10.1172/JCI43578
    • (2011) J Clin Invest , vol.121 , Issue.7 , pp. 2833-2844
    • Cao, K.1    Blair, C.D.2    Faddah, D.A.3    Kieckhaefer, J.E.4    Olive, M.5    Erdos, M.R.6    Nabel, E.G.7    Collins, F.S.8
  • 21
    • 79959776029 scopus 로고    scopus 로고
    • Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson–Gilford progeria syndrome cells
    • Cao K, Graziotto JJ, Blair CD, Mazzulli JR, Erdos MR, Krainc D, Collins FS (2011b) Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson–Gilford progeria syndrome cells. Sci Transl Med. doi:10.1126/scitranslmed.3002346
    • (2011) Sci Transl Med
    • Cao, K.1    Graziotto, J.J.2    Blair, C.D.3    Mazzulli, J.R.4    Erdos, M.R.5    Krainc, D.6    Collins, F.S.7
  • 24
    • 0037342243 scopus 로고    scopus 로고
    • A new clinical condition linked to a novel mutation in lamins A and C with generalized lipoatrophy, insulin-resistant diabetes, disseminated leukomelanodermic papules, liver steatosis, and cardiomyopathy
    • COI: 1:CAS:528:DC%2BD3sXit1egtbg%3D, PID: 12629077
    • Caux F, Dubosclard E, Lascols O, Buendia B, Chazouilleres O, Cohen A, Courvalin JC, Laroche L, Capeau J, Vigouroux C, Christin-Maitre S (2003) A new clinical condition linked to a novel mutation in lamins A and C with generalized lipoatrophy, insulin-resistant diabetes, disseminated leukomelanodermic papules, liver steatosis, and cardiomyopathy. J Clin Endocrinol Metab 88(3):1006–1013. doi:10.1210/jc.2002-021506
    • (2003) J Clin Endocrinol Metab , vol.88 , Issue.3 , pp. 1006-1013
    • Caux, F.1    Dubosclard, E.2    Lascols, O.3    Buendia, B.4    Chazouilleres, O.5    Cohen, A.6    Courvalin, J.C.7    Laroche, L.8    Capeau, J.9    Vigouroux, C.10    Christin-Maitre, S.11
  • 27
    • 84898887269 scopus 로고    scopus 로고
    • Dysregulated interactions between lamin A and SUN1 induce abnormalities in the nuclear envelope and endoplasmic reticulum in progeric laminopathies
    • COI: 1:CAS:528:DC%2BC2cXos1Churo%3D, PID: 24522183
    • Chen ZJ, Wang WP, Chen YC, Wang JY, Lin WH, Tai LA, Liou GG, Yang CS, Chi YH (2014) Dysregulated interactions between lamin A and SUN1 induce abnormalities in the nuclear envelope and endoplasmic reticulum in progeric laminopathies. J Cell Sci 127(Pt 8):1792–1804. doi:10.1242/jcs.139683
    • (2014) J Cell Sci , vol.127 , pp. 1792-1804
    • Chen, Z.J.1    Wang, W.P.2    Chen, Y.C.3    Wang, J.Y.4    Lin, W.H.5    Tai, L.A.6    Liou, G.G.7    Yang, C.S.8    Chi, Y.H.9
  • 29
    • 0025943935 scopus 로고
    • Altered response of progeria fibroblasts to epidermal growth factor
    • COI: 1:CAS:528:DyaK38Xht1Khs7o%3D, PID: 1808212
    • Colige A, Nusgens B, Lapiere CM (1991) Altered response of progeria fibroblasts to epidermal growth factor. J Cell Sci 100(Pt 3):649–655
    • (1991) J Cell Sci , vol.100 , pp. 649-655
    • Colige, A.1    Nusgens, B.2    Lapiere, C.M.3
  • 30
    • 34447543913 scopus 로고    scopus 로고
    • Cellular senescence in cancer and aging
    • COI: 1:CAS:528:DC%2BD2sXovFaltrc%3D, PID: 17662938
    • Collado M, Blasco MA, Serrano M (2007) Cellular senescence in cancer and aging. Cell 130(2):223–233. doi:10.1016/j.cell.2007.07.003
    • (2007) Cell , vol.130 , Issue.2 , pp. 223-233
    • Collado, M.1    Blasco, M.A.2    Serrano, M.3
  • 31
    • 28344445866 scopus 로고    scopus 로고
    • Rescue of heterochromatin organization in Hutchinson–Gilford progeria by drug treatment
    • COI: 1:CAS:528:DC%2BD2MXhtlSms7vF, PID: 16261260
    • Columbaro M, Capanni C, Mattioli E, Novelli G, Parnaik VK, Squarzoni S, Maraldi NM, Lattanzi G (2005) Rescue of heterochromatin organization in Hutchinson–Gilford progeria by drug treatment. Cell Mol Life Sci 62(22):2669–2678. doi:10.1007/s00018-005-5318-6
    • (2005) Cell Mol Life Sci , vol.62 , Issue.22 , pp. 2669-2678
    • Columbaro, M.1    Capanni, C.2    Mattioli, E.3    Novelli, G.4    Parnaik, V.K.5    Squarzoni, S.6    Maraldi, N.M.7    Lattanzi, G.8
  • 32
    • 0035694907 scopus 로고    scopus 로고
    • Intermediate filaments at a glance
    • COI: 1:CAS:528:DC%2BD38XnvFKmsQ%3D%3D, PID: 11792800
    • Coulombe PA, Ma L, Yamada S, Wawersik M (2001) Intermediate filaments at a glance. J Cell Sci 114(Pt 24):4345–4347
    • (2001) J Cell Sci , vol.114 , pp. 4345-4347
    • Coulombe, P.A.1    Ma, L.2    Yamada, S.3    Wawersik, M.4
  • 33
    • 1942469525 scopus 로고    scopus 로고
    • Novel lamin A/C gene (LMNA) mutations in atypical progeroid syndromes
    • COI: 1:CAS:528:DC%2BD2cXktl2ktrg%3D, PID: 15060110
    • Csoka AB, Cao H, Sammak PJ, Constantinescu D, Schatten GP, Hegele RA (2004) Novel lamin A/C gene (LMNA) mutations in atypical progeroid syndromes. J Med Genet 41(4):304–308
    • (2004) J Med Genet , vol.41 , Issue.4 , pp. 304-308
    • Csoka, A.B.1    Cao, H.2    Sammak, P.J.3    Constantinescu, D.4    Schatten, G.P.5    Hegele, R.A.6
  • 34
    • 33745904741 scopus 로고    scopus 로고
    • Distinct structural and mechanical properties of the nuclear lamina in Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BD28XntlKkur8%3D, PID: 16801550
    • Dahl KN, Scaffidi P, Islam MF, Yodh AG, Wilson KL, Misteli T (2006) Distinct structural and mechanical properties of the nuclear lamina in Hutchinson–Gilford progeria syndrome. Proc Natl Acad Sci USA 103(27):10271–10276. doi:10.1073/pnas.0601058103
    • (2006) Proc Natl Acad Sci USA , vol.103 , Issue.27 , pp. 10271-10276
    • Dahl, K.N.1    Scaffidi, P.2    Islam, M.F.3    Yodh, A.G.4    Wilson, K.L.5    Misteli, T.6
  • 35
    • 70350236483 scopus 로고    scopus 로고
    • The posttranslational processing of prelamin A and disease
    • COI: 1:CAS:528:DC%2BD1MXht12qsrvP, PID: 19453251
    • Davies BSJ, Fong LG, Yang SH, Coffinier C, Young SG (2009) The posttranslational processing of prelamin A and disease. Annu Rev Genomics Hum Genet 10:153–174. doi:10.1146/annurev-genom-082908-150150
    • (2009) Annu Rev Genomics Hum Genet , vol.10 , pp. 153-174
    • Davies, B.S.J.1    Fong, L.G.2    Yang, S.H.3    Coffinier, C.4    Young, S.G.5
  • 39
    • 0033763071 scopus 로고    scopus 로고
    • Lamina-associated polypeptide 2α binds intranuclear A-type lamins
    • COI: 1:CAS:528:DC%2BD3cXnvF2gtrk%3D, PID: 10984438
    • Dechat T, Korbei B, Vaughan OA, Vlcek S, Hutchison CJ, Foisner R (2000) Lamina-associated polypeptide 2α binds intranuclear A-type lamins. J Cell Sci 113(Pt 19):3473–3484
    • (2000) J Cell Sci , vol.113 , pp. 3473-3484
    • Dechat, T.1    Korbei, B.2    Vaughan, O.A.3    Vlcek, S.4    Hutchison, C.J.5    Foisner, R.6
  • 40
    • 34247383902 scopus 로고    scopus 로고
    • Alterations in mitosis and cell cycle progression caused by a mutant lamin A known to accelerate human aging
    • COI: 1:CAS:528:DC%2BD2sXjvFKiur4%3D, PID: 17360326
    • Dechat T, Shimi T, Adam SA, Rusinol AE, Andres DA, Spielmann HP, Sinensky MS, Goldman RD (2007) Alterations in mitosis and cell cycle progression caused by a mutant lamin A known to accelerate human aging. Proc Natl Acad Sci USA 104(12):4955–4960. doi:10.1073/pnas.0700854104
    • (2007) Proc Natl Acad Sci USA , vol.104 , Issue.12 , pp. 4955-4960
    • Dechat, T.1    Shimi, T.2    Adam, S.A.3    Rusinol, A.E.4    Andres, D.A.5    Spielmann, H.P.6    Sinensky, M.S.7    Goldman, R.D.8
  • 41
    • 66449126030 scopus 로고    scopus 로고
    • Nuclear lamins and chromatin: when structure meets function
    • COI: 1:CAS:528:DC%2BD1MXpsFehs7g%3D, PID: 19154754
    • Dechat T, Adam SA, Goldman RD (2009) Nuclear lamins and chromatin: when structure meets function. Adv Enzyme Regul 49(1):157–166. doi:10.1016/j.advenzreg.2008.12.003
    • (2009) Adv Enzyme Regul , vol.49 , Issue.1 , pp. 157-166
    • Dechat, T.1    Adam, S.A.2    Goldman, R.D.3
  • 43
    • 80052417470 scopus 로고    scopus 로고
    • Lamina-independent lamins in the nuclear interior serve important functions
    • COI: 1:CAS:528:DC%2BC38XnsVOju74%3D, PID: 21209392
    • Dechat T, Gesson K, Foisner R (2010b) Lamina-independent lamins in the nuclear interior serve important functions. Cold Spring Harb Symp Quant Biol 75:533–543. doi:10.1101/sqb.2010.75.018
    • (2010) Cold Spring Harb Symp Quant Biol , vol.75 , pp. 533-543
    • Dechat, T.1    Gesson, K.2    Foisner, R.3
  • 44
    • 67349126486 scopus 로고    scopus 로고
    • Telomere length in Hutchinson–Gilford Progeria Syndrome
    • COI: 1:CAS:528:DC%2BD1MXls1ynsbg%3D, PID: 19428457
    • Decker ML, Chavez E, Vulto I, Lansdorp PM (2009) Telomere length in Hutchinson–Gilford Progeria Syndrome. Mech Ageing Dev 130(6):377–383. doi:10.1016/j.mad.2009.03.001
    • (2009) Mech Ageing Dev , vol.130 , Issue.6 , pp. 377-383
    • Decker, M.L.1    Chavez, E.2    Vulto, I.3    Lansdorp, P.M.4
  • 45
    • 33645116709 scopus 로고    scopus 로고
    • The truncated prelamin A in Hutchinson–Gilford progeria syndrome alters segregation of A-type and B-type lamin homopolymers
    • COI: 1:CAS:528:DC%2BD28Xis1Siu7g%3D, PID: 16481358
    • Delbarre E, Tramier M, Coppey-Moisan M, Gaillard C, Courvalin JC, Buendia B (2006) The truncated prelamin A in Hutchinson–Gilford progeria syndrome alters segregation of A-type and B-type lamin homopolymers. Hum Mol Genet 15(7):1113–1122. doi:10.1093/hmg/ddl026
    • (2006) Hum Mol Genet , vol.15 , Issue.7 , pp. 1113-1122
    • Delbarre, E.1    Tramier, M.2    Coppey-Moisan, M.3    Gaillard, C.4    Courvalin, J.C.5    Buendia, B.6
  • 47
    • 79957654428 scopus 로고    scopus 로고
    • The lamin protein family
    • PID: 21639948
    • Dittmer T, Misteli T (2011) The lamin protein family. Genome Biol. doi:10.1186/Gb-2011-12-5-222
    • (2011) Genome Biol
    • Dittmer, T.1    Misteli, T.2
  • 48
    • 64249101119 scopus 로고    scopus 로고
    • Novel LMNA gene mutation in a patient with Atypical Werner’s Syndrome
    • COI: 1:CAS:528:DC%2BD1MXktlGltb8%3D, PID: 19270485
    • Doh YJ, Kim HK, Jung ED, Choi SH, Kim JG, Kim BW, Lee IK (2009) Novel LMNA gene mutation in a patient with Atypical Werner’s Syndrome. Korean J Intern Med 24(1):68–72. doi:10.3904/kjim.2009.24.1.68
    • (2009) Korean J Intern Med , vol.24 , Issue.1 , pp. 68-72
    • Doh, Y.J.1    Kim, H.K.2    Jung, E.D.3    Choi, S.H.4    Kim, J.G.5    Kim, B.W.6    Lee, I.K.7
  • 49
    • 33645743546 scopus 로고    scopus 로고
    • Lamina-associated polypeptide 2α regulates cell cycle progression and differentiation via the retinoblastoma-E2F pathway
    • COI: 1:CAS:528:DC%2BD28XjsFCgt7Y%3D, PID: 16606692
    • Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotzmann J, Hutchison CJ, Foisner R (2006) Lamina-associated polypeptide 2α regulates cell cycle progression and differentiation via the retinoblastoma-E2F pathway. J Cell Biol 173(1):83–93. doi:10.1083/jcb.200511149
    • (2006) J Cell Biol , vol.173 , Issue.1 , pp. 83-93
    • Dorner, D.1    Vlcek, S.2    Foeger, N.3    Gajewski, A.4    Makolm, C.5    Gotzmann, J.6    Hutchison, C.J.7    Foisner, R.8
  • 51
    • 84856646656 scopus 로고    scopus 로고
    • Accelerated aging syndromes, are they relevant to normal human aging?
    • COI: 1:CAS:528:DC%2BC3MXhsFCht7fI
    • Dreesen O, Stewart CL (2011) Accelerated aging syndromes, are they relevant to normal human aging? Aging (Albany NY) 3(9):889–895
    • (2011) Aging (Albany NY) , vol.3 , Issue.9 , pp. 889-895
    • Dreesen, O.1    Stewart, C.L.2
  • 52
    • 84873531288 scopus 로고    scopus 로고
    • Lamin A/C is expressed in pluripotent mouse embryonic stem cells
    • PID: 23324457
    • Eckersley-Maslin MA, Bergmann JH, Lazar Z, Spector DL (2013) Lamin A/C is expressed in pluripotent mouse embryonic stem cells. Nucleus 4(1):53–60. doi:10.4161/nucl.23384
    • (2013) Nucleus , vol.4 , Issue.1 , pp. 53-60
    • Eckersley-Maslin, M.A.1    Bergmann, J.H.2    Lazar, Z.3    Spector, D.L.4
  • 55
    • 33645060977 scopus 로고    scopus 로고
    • A protein farnesyltransferase inhibitor ameliorates disease in a mouse model of progeria
    • COI: 1:CAS:528:DC%2BD28XitlSnsb8%3D, PID: 16484451
    • Fong LG, Frost D, Meta M, Qiao X, Yang SH, Coffinier C, Young SG (2006) A protein farnesyltransferase inhibitor ameliorates disease in a mouse model of progeria. Science 311(5767):1621–1623. doi:10.1126/science.1124875
    • (2006) Science , vol.311 , Issue.5767 , pp. 1621-1623
    • Fong, L.G.1    Frost, D.2    Meta, M.3    Qiao, X.4    Yang, S.H.5    Coffinier, C.6    Young, S.G.7
  • 56
    • 84922661954 scopus 로고    scopus 로고
    • Sulforaphane enhances progerin clearance in Hutchinson–Gilford progeria fibroblasts
    • COI: 1:CAS:528:DC%2BC2MXhslantbw%3D, PID: 25510262
    • Gabriel D, Roedl D, Gordon LB, Djabali K (2015) Sulforaphane enhances progerin clearance in Hutchinson–Gilford progeria fibroblasts. Aging Cell 14(1):78–91. doi:10.1111/acel.12300
    • (2015) Aging Cell , vol.14 , Issue.1 , pp. 78-91
    • Gabriel, D.1    Roedl, D.2    Gordon, L.B.3    Djabali, K.4
  • 58
    • 84903541437 scopus 로고    scopus 로고
    • Extracellular matrix: a dynamic microenvironment for stem cell niche
    • COI: 1:CAS:528:DC%2BC2cXhs1Grtro%3D, PID: 24418517
    • Gattazzo F, Urciuolo A, Bonaldo P (2014) Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta 1840(8):2506–2519. doi:10.1016/j.bbagen.2014.01.010
    • (2014) Biochim Biophys Acta , vol.1840 , Issue.8 , pp. 2506-2519
    • Gattazzo, F.1    Urciuolo, A.2    Bonaldo, P.3
  • 59
    • 84901473315 scopus 로고    scopus 로고
    • Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease
    • COI: 1:CAS:528:DC%2BC2cXovFCiu7k%3D, PID: 24374133
    • Gesson K, Vidak S, Foisner R (2014) Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease. Semin Cell Dev Biol 29:116–124. doi:10.1016/j.semcdb.2013.12.009
    • (2014) Semin Cell Dev Biol , vol.29 , pp. 116-124
    • Gesson, K.1    Vidak, S.2    Foisner, R.3
  • 60
    • 54949147176 scopus 로고    scopus 로고
    • New regulators of NF-κB in inflammation
    • COI: 1:CAS:528:DC%2BD1cXht12isLfN, PID: 18927578
    • Ghosh S, Hayden MS (2008) New regulators of NF-κB in inflammation. Nat Rev Immunol 8(11):837–848. doi:10.1038/nri2423
    • (2008) Nat Rev Immunol , vol.8 , Issue.11 , pp. 837-848
    • Ghosh, S.1    Hayden, M.S.2
  • 61
    • 84903827705 scopus 로고    scopus 로고
    • Genetics of aging, progeria and lamin disorders
    • COI: 1:CAS:528:DC%2BC2cXht12nu73F, PID: 25005744
    • Ghosh S, Zhou Z (2014) Genetics of aging, progeria and lamin disorders. Curr Opin Genet Dev 26:41–46. doi:10.1016/j.gde.2014.05.003
    • (2014) Curr Opin Genet Dev , vol.26 , pp. 41-46
    • Ghosh, S.1    Zhou, Z.2
  • 63
    • 58149186768 scopus 로고    scopus 로고
    • Fast regulation of AP-1 activity through interaction of lamin A/C, ERK1/2, and c-Fos at the nuclear envelope
    • COI: 1:CAS:528:DC%2BD1cXhsVartbjE, PID: 19015316
    • Gonzalez JM, Navarro-Puche A, Casar B, Crespo P, Andres V (2008) Fast regulation of AP-1 activity through interaction of lamin A/C, ERK1/2, and c-Fos at the nuclear envelope. J Cell Biol 183(4):653–666. doi:10.1083/jcb.200805049
    • (2008) J Cell Biol , vol.183 , Issue.4 , pp. 653-666
    • Gonzalez, J.M.1    Navarro-Puche, A.2    Casar, B.3    Crespo, P.4    Andres, V.5
  • 64
    • 77957663987 scopus 로고    scopus 로고
    • Nurturing the genome: A-type lamins preserve genomic stability
    • PID: 21326943
    • Gonzalez-Suarez I, Gonzalo S (2010) Nurturing the genome: A-type lamins preserve genomic stability. Nucleus 1(2):129–135. doi:10.4161/nucl.1.2.10797
    • (2010) Nucleus , vol.1 , Issue.2 , pp. 129-135
    • Gonzalez-Suarez, I.1    Gonzalo, S.2
  • 65
    • 84931265120 scopus 로고    scopus 로고
    • DNA repair defects and genome instability in Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BC2MXhtVWqsrvL, PID: 26079711
    • Gonzalo S, Kreienkamp R (2015) DNA repair defects and genome instability in Hutchinson–Gilford progeria syndrome. Curr Opin Cell Biol 34:75–83. doi:10.1016/j.ceb.2015.05.007
    • (2015) Curr Opin Cell Biol , vol.34 , pp. 75-83
    • Gonzalo, S.1    Kreienkamp, R.2
  • 68
    • 84893431595 scopus 로고    scopus 로고
    • Progeria: A paradigm for translational medicine
    • COI: 1:CAS:528:DC%2BC2cXhs1Gku7s%3D, PID: 24485450
    • Gordon LB, Rothman FG, Lopez-Otin C, Misteli T (2014) Progeria: A paradigm for translational medicine. Cell 156(3):400–407. doi:10.1016/j.cell.2013.12.028
    • (2014) Cell , vol.156 , Issue.3 , pp. 400-407
    • Gordon, L.B.1    Rothman, F.G.2    Lopez-Otin, C.3    Misteli, T.4
  • 69
    • 84961624556 scopus 로고    scopus 로고
    • Lamins and emerin in muscular distrophy
    • Winder SJ, (ed), Eureka/Landes Bioscience, New Yoork
    • Gotzman J, Foisner R (2006) Lamins and emerin in muscular distrophy. In: Winder SJ (ed) Molecular Mechanisms of Muscular Distrophy. Eureka/Landes Bioscience, New Yoork, pp 142–159
    • (2006) Molecular Mechanisms of Muscular Distrophy , pp. 142-159
    • Gotzman, J.1    Foisner, R.2
  • 70
    • 84930719986 scopus 로고    scopus 로고
    • Lamins: nuclear intermediate filament proteins with fundamental functions in nuclear mechanics and genome regulation
    • COI: 1:CAS:528:DC%2BC2MXhsVyrt7%2FP, PID: 25747401
    • Gruenbaum Y, Foisner R (2015) Lamins: nuclear intermediate filament proteins with fundamental functions in nuclear mechanics and genome regulation. Annu Rev Biochem 84:131–164. doi:10.1146/annurev-biochem-060614-034115
    • (2015) Annu Rev Biochem , vol.84 , pp. 131-164
    • Gruenbaum, Y.1    Foisner, R.2
  • 71
    • 34047113394 scopus 로고    scopus 로고
    • Progeria of stem cells: stem cell exhaustion in Hutchinson–Gilford progeria syndrome
    • PID: 17301031
    • Halaschek-Wiener J, Brooks-Wilson A (2007) Progeria of stem cells: stem cell exhaustion in Hutchinson–Gilford progeria syndrome. J Gerontol A Biol Sci Med Sci 62(1):3–8
    • (2007) J Gerontol A Biol Sci Med Sci , vol.62 , Issue.1 , pp. 3-8
    • Halaschek-Wiener, J.1    Brooks-Wilson, A.2
  • 72
    • 75149142796 scopus 로고    scopus 로고
    • Differential requirement of CAAX-mediated posttranslational processing for Rheb localization and signaling
    • COI: 1:CAS:528:DC%2BD1MXht1OgsrzP, PID: 19838215
    • Hanker AB, Mitin N, Wilder RS, Henske EP, Tamanoi F, Cox AD, Der CJ (2010) Differential requirement of CAAX-mediated posttranslational processing for Rheb localization and signaling. Oncogene 29(3):380–391. doi:10.1038/onc.2009.336
    • (2010) Oncogene , vol.29 , Issue.3 , pp. 380-391
    • Hanker, A.B.1    Mitin, N.2    Wilder, R.S.3    Henske, E.P.4    Tamanoi, F.5    Cox, A.D.6    Der, C.J.7
  • 75
    • 34250880140 scopus 로고    scopus 로고
    • Intermediate filaments: from cell architecture to nanomechanics
    • COI: 1:CAS:528:DC%2BD2sXmvVaktLg%3D, PID: 17551517
    • Herrmann H, Bar H, Kreplak L, Strelkov SV, Aebi U (2007) Intermediate filaments: from cell architecture to nanomechanics. Nat Rev Mol Cell Biol 8(7):562–573. doi:10.1038/nrm2197
    • (2007) Nat Rev Mol Cell Biol , vol.8 , Issue.7 , pp. 562-573
    • Herrmann, H.1    Bar, H.2    Kreplak, L.3    Strelkov, S.V.4    Aebi, U.5
  • 76
    • 84862882928 scopus 로고    scopus 로고
    • Lamins at a glance
    • COI: 1:CAS:528:DC%2BC38XhtVGltr7I, PID: 22669459
    • Ho CY, Lammerding J (2012) Lamins at a glance. J Cell Sci 125(9):2087–2093. doi:10.1242/Jcs.087288
    • (2012) J Cell Sci , vol.125 , Issue.9 , pp. 2087-2093
    • Ho, C.Y.1    Lammerding, J.2
  • 77
    • 70349859881 scopus 로고    scopus 로고
    • DNA damage, aging, and cancer
    • COI: 1:CAS:528:DC%2BD1MXht1GiurbK, PID: 19812404
    • Hoeijmakers JH (2009) DNA damage, aging, and cancer. N Engl J Med 361(15):1475–1485. doi:10.1056/NEJMra0804615
    • (2009) N Engl J Med , vol.361 , Issue.15 , pp. 1475-1485
    • Hoeijmakers, J.H.1
  • 78
    • 84879870498 scopus 로고    scopus 로고
    • Notch signaling at a glance
    • COI: 1:CAS:528:DC%2BC3sXhtFaks7bN, PID: 23729744
    • Hori K, Sen A, Artavanis-Tsakonas S (2013) Notch signaling at a glance. J Cell Sci 126(Pt 10):2135–2140. doi:10.1242/jcs.127308
    • (2013) J Cell Sci , vol.126 , pp. 2135-2140
    • Hori, K.1    Sen, A.2    Artavanis-Tsakonas, S.3
  • 80
    • 84922128722 scopus 로고    scopus 로고
    • Mechanotransduction and extracellular matrix homeostasis
    • COI: 1:CAS:528:DC%2BC2cXhvVCnu7%2FJ, PID: 25355505
    • Humphrey JD, Dufresne ER, Schwartz MA (2014) Mechanotransduction and extracellular matrix homeostasis. Nat Rev Mol Cell Biol 15(12):802–812. doi:10.1038/nrm3896
    • (2014) Nat Rev Mol Cell Biol , vol.15 , Issue.12 , pp. 802-812
    • Humphrey, J.D.1    Dufresne, E.R.2    Schwartz, M.A.3
  • 82
    • 28744453386 scopus 로고    scopus 로고
    • Phenotypic heterogeneity in body fat distribution in patients with atypical Werner’s Syndrome due to heterozygous Arg133Leu lamin A/C mutation
    • COI: 1:CAS:528:DC%2BD2MXhtlaltLvI, PID: 16174718
    • Jacob KN, Baptista F, dos Santos HG, Oshima J, Agarwal AK, Garg A (2005) Phenotypic heterogeneity in body fat distribution in patients with atypical Werner’s Syndrome due to heterozygous Arg133Leu lamin A/C mutation. J Clin Endocrinol Metab 90(12):6699–6706. doi:10.1210/jc.2005-0939
    • (2005) J Clin Endocrinol Metab , vol.90 , Issue.12 , pp. 6699-6706
    • Jacob, K.N.1    Baptista, F.2    dos Santos, H.G.3    Oshima, J.4    Agarwal, A.K.5    Garg, A.6
  • 83
    • 3042829496 scopus 로고    scopus 로고
    • A-type lamins regulate retinoblastoma protein function by promoting subnuclear localization and preventing proteasomal degradation
    • COI: 1:CAS:528:DC%2BD2cXlvVahsbc%3D, PID: 15210943
    • Johnson BR, Nitta RT, Frock RL, Mounkes L, Barbie DA, Stewart CL, Harlow E, Kennedy BK (2004) A-type lamins regulate retinoblastoma protein function by promoting subnuclear localization and preventing proteasomal degradation. Proc Natl Acad Sci USA 101(26):9677–9682. doi:10.1073/pnas.0403250101
    • (2004) Proc Natl Acad Sci USA , vol.101 , Issue.26 , pp. 9677-9682
    • Johnson, B.R.1    Nitta, R.T.2    Frock, R.L.3    Mounkes, L.4    Barbie, D.A.5    Stewart, C.L.6    Harlow, E.7    Kennedy, B.K.8
  • 84
    • 84872527628 scopus 로고    scopus 로고
    • mTOR is a key modulator of ageing and age-related disease
    • COI: 1:CAS:528:DC%2BC3sXptlyntg%3D%3D, PID: 23325216
    • Johnson SC, Rabinovitch PS, Kaeberlein M (2013) mTOR is a key modulator of ageing and age-related disease. Nature 493(7432):338–345. doi:10.1038/nature11861
    • (2013) Nature , vol.493 , Issue.7432 , pp. 338-345
    • Johnson, S.C.1    Rabinovitch, P.S.2    Kaeberlein, M.3
  • 85
    • 77950501014 scopus 로고    scopus 로고
    • mTOR regulation of autophagy
    • COI: 1:CAS:528:DC%2BC3cXjs12nu74%3D, PID: 20083114
    • Jung CH, Ro SH, Cao J, Otto NM, Kim DH (2010) mTOR regulation of autophagy. FEBS Lett 584(7):1287–1295. doi:10.1016/j.febslet.2010.01.017
    • (2010) FEBS Lett , vol.584 , Issue.7 , pp. 1287-1295
    • Jung, C.H.1    Ro, S.H.2    Cao, J.3    Otto, N.M.4    Kim, D.H.5
  • 86
    • 84879462736 scopus 로고    scopus 로고
    • LMNA-associated cardiocutaneous progeria: an inherited autosomal dominant premature aging syndrome with late onset
    • PID: 23666920
    • Kane MS, Lindsay ME, Judge DP, Barrowman J, Ap Rhys C, Simonson L, Dietz HC, Michaelis S (2013) LMNA-associated cardiocutaneous progeria: an inherited autosomal dominant premature aging syndrome with late onset. Am J Med Genet A 161A(7):1599–1611. doi:10.1002/ajmg.a.35971
    • (2013) Am J Med Genet A , vol.161A , Issue.7 , pp. 1599-1611
    • Kane, M.S.1    Lindsay, M.E.2    Judge, D.P.3    Barrowman, J.4    Ap Rhys, C.5    Simonson, L.6    Dietz, H.C.7    Michaelis, S.8
  • 88
    • 35148898709 scopus 로고    scopus 로고
    • New approaches to progeria
    • PID: 17908771
    • Kieran MW, Gordon L, Kleinman M (2007) New approaches to progeria. Pediatrics 120(4):834–841. doi:10.1542/peds.2007-1356
    • (2007) Pediatrics , vol.120 , Issue.4 , pp. 834-841
    • Kieran, M.W.1    Gordon, L.2    Kleinman, M.3
  • 91
    • 80054715001 scopus 로고    scopus 로고
    • Lamin A and lamin C form homodimers and coexist in higher complex forms both in the nucleoplasmic fraction and in the lamina of cultured human cells
    • PID: 22033280
    • Kolb T, Maass K, Hergt M, Aebi U, Herrmann H (2011) Lamin A and lamin C form homodimers and coexist in higher complex forms both in the nucleoplasmic fraction and in the lamina of cultured human cells. Nucleus 2(5):425–433. doi:10.4161/nucl.2.5.17765
    • (2011) Nucleus , vol.2 , Issue.5 , pp. 425-433
    • Kolb, T.1    Maass, K.2    Hergt, M.3    Aebi, U.4    Herrmann, H.5
  • 93
    • 78549252392 scopus 로고    scopus 로고
    • Identification of differential protein interactors of lamin A and progerin
    • PID: 21327095
    • Kubben N, Voncken JW, Demmers J, Calis C, van Almen G, Pinto Y, Misteli T (2010a) Identification of differential protein interactors of lamin A and progerin. Nucleus 1(6):513–525. doi:10.4161/nucl.1.6.13512
    • (2010) Nucleus , vol.1 , Issue.6 , pp. 513-525
    • Kubben, N.1    Voncken, J.W.2    Demmers, J.3    Calis, C.4    van Almen, G.5    Pinto, Y.6    Misteli, T.7
  • 94
    • 78549295171 scopus 로고    scopus 로고
    • Mapping of protein- and chromatin-interactions at the nuclear lamina
    • PID: 21327087
    • Kubben N, Voncken JW, Misteli T (2010b) Mapping of protein- and chromatin-interactions at the nuclear lamina. Nucleus 1(6):460–471. doi:10.4161/nucl.1.6.13513
    • (2010) Nucleus , vol.1 , Issue.6 , pp. 460-471
    • Kubben, N.1    Voncken, J.W.2    Misteli, T.3
  • 95
    • 84867657161 scopus 로고    scopus 로고
    • Mapping of lamin A- and progerin-interacting genome regions
    • COI: 1:CAS:528:DC%2BC38XhtlOlurvF, PID: 22610065
    • Kubben N, Adriaens M, Meuleman W, Voncken JW, van Steensel B, Misteli T (2012) Mapping of lamin A- and progerin-interacting genome regions. Chromosoma 121(5):447–464. doi:10.1007/s00412-012-0376-7
    • (2012) Chromosoma , vol.121 , Issue.5 , pp. 447-464
    • Kubben, N.1    Adriaens, M.2    Meuleman, W.3    Voncken, J.W.4    van Steensel, B.5    Misteli, T.6
  • 96
    • 84958174324 scopus 로고    scopus 로고
    • A high-content imaging-based screening pipeline for the systematic identification of anti-progeroid compounds
    • PID: 26341717
    • Kubben N, Brimacombe KR, Donegan M, Li Z, Misteli T (2015) A high-content imaging-based screening pipeline for the systematic identification of anti-progeroid compounds. Methods. doi:10.1016/j.ymeth.2015.08.024
    • (2015) Methods
    • Kubben, N.1    Brimacombe, K.R.2    Donegan, M.3    Li, Z.4    Misteli, T.5
  • 97
    • 34247390746 scopus 로고    scopus 로고
    • Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases
    • COI: 1:CAS:528:DC%2BD2sXksFSjsr8%3D, PID: 17450177
    • Kudlow BA, Kennedy BK, Monnat RJ Jr (2007) Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat Rev Mol Cell Biol 8(5):394–404. doi:10.1038/nrm2161
    • (2007) Nat Rev Mol Cell Biol , vol.8 , Issue.5 , pp. 394-404
    • Kudlow, B.A.1    Kennedy, B.K.2    Monnat, R.J.3
  • 99
    • 53249121556 scopus 로고    scopus 로고
    • Sirtuins–novel therapeutic targets to treat age-associated diseases
    • COI: 1:CAS:528:DC%2BD1cXhtFOlu7rI, PID: 18827827
    • Lavu S, Boss O, Elliott PJ, Lambert PD (2008) Sirtuins–novel therapeutic targets to treat age-associated diseases. Nat Rev Drug Discov 7(10):841–853. doi:10.1038/nrd2665
    • (2008) Nat Rev Drug Discov , vol.7 , Issue.10 , pp. 841-853
    • Lavu, S.1    Boss, O.2    Elliott, P.J.3    Lambert, P.D.4
  • 101
    • 84870506099 scopus 로고    scopus 로고
    • Resveratrol rescues SIRT1-dependent adult stem cell decline and alleviates progeroid features in laminopathy-based progeria
    • COI: 1:CAS:528:DC%2BC38XhslyntbnN, PID: 23217256
    • Liu B, Ghosh S, Yang X, Zheng H, Liu X, Wang Z, Jin G, Zheng B, Kennedy BK, Suh Y, Kaeberlein M, Tryggvason K, Zhou Z (2012) Resveratrol rescues SIRT1-dependent adult stem cell decline and alleviates progeroid features in laminopathy-based progeria. Cell Metab 16(6):738–750. doi:10.1016/j.cmet.2012.11.007
    • (2012) Cell Metab , vol.16 , Issue.6 , pp. 738-750
    • Liu, B.1    Ghosh, S.2    Yang, X.3    Zheng, H.4    Liu, X.5    Wang, Z.6    Jin, G.7    Zheng, B.8    Kennedy, B.K.9    Suh, Y.10    Kaeberlein, M.11    Tryggvason, K.12    Zhou, Z.13
  • 102
    • 84878738557 scopus 로고    scopus 로고
    • Depleting the methyltransferase Suv39h1 improves DNA repair and extends lifespan in a progeria mouse model
    • PID: 23695662
    • Liu B, Wang Z, Zhang L, Ghosh S, Zheng H, Zhou Z (2013) Depleting the methyltransferase Suv39h1 improves DNA repair and extends lifespan in a progeria mouse model. Nat Commun 4:1868. doi:10.1038/ncomms2885
    • (2013) Nat Commun , vol.4 , pp. 1868
    • Liu, B.1    Wang, Z.2    Zhang, L.3    Ghosh, S.4    Zheng, H.5    Zhou, Z.6
  • 103
    • 84878864199 scopus 로고    scopus 로고
    • The hallmarks of aging
    • COI: 1:CAS:528:DC%2BC3sXptFKms78%3D, PID: 23746838
    • Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153(6):1194–1217. doi:10.1016/j.cell.2013.05.039
    • (2013) Cell , vol.153 , Issue.6 , pp. 1194-1217
    • Lopez-Otin, C.1    Blasco, M.A.2    Partridge, L.3    Serrano, M.4    Kroemer, G.5
  • 104
    • 84885033978 scopus 로고    scopus 로고
    • Lamin A/C-promoter interactions specify chromatin state-dependent transcription outcomes
    • COI: 1:CAS:528:DC%2BC3sXhs1eltr%2FE, PID: 23861385
    • Lund E, Oldenburg AR, Delbarre E, Freberg CT, Duband-Goulet I, Eskeland R, Buendia B, Collas P (2013) Lamin A/C-promoter interactions specify chromatin state-dependent transcription outcomes. Genome Res 23(10):1580–1589. doi:10.1101/gr.159400.113
    • (2013) Genome Res , vol.23 , Issue.10 , pp. 1580-1589
    • Lund, E.1    Oldenburg, A.R.2    Delbarre, E.3    Freberg, C.T.4    Duband-Goulet, I.5    Eskeland, R.6    Buendia, B.7    Collas, P.8
  • 105
    • 84924233960 scopus 로고    scopus 로고
    • Distinct features of lamin A-interacting chromatin domains mapped by ChIP-sequencing from sonicated or micrococcal nuclease-digested chromatin
    • PID: 25602132
    • Lund EG, Duband-Goulet I, Oldenburg A, Buendia B, Collas P (2015) Distinct features of lamin A-interacting chromatin domains mapped by ChIP-sequencing from sonicated or micrococcal nuclease-digested chromatin. Nucleus 6(1):30–39. doi:10.4161/19491034.2014.990855
    • (2015) Nucleus , vol.6 , Issue.1 , pp. 30-39
    • Lund, E.G.1    Duband-Goulet, I.2    Oldenburg, A.3    Buendia, B.4    Collas, P.5
  • 106
    • 77956420489 scopus 로고    scopus 로고
    • Can autophagy promote longevity?
    • COI: 1:CAS:528:DC%2BC3cXhtFSju7zO, PID: 20811357
    • Madeo F, Tavernarakis N, Kroemer G (2010) Can autophagy promote longevity? Nat Cell Biol 12(9):842–846. doi:10.1038/ncb0910-842
    • (2010) Nat Cell Biol , vol.12 , Issue.9 , pp. 842-846
    • Madeo, F.1    Tavernarakis, N.2    Kroemer, G.3
  • 107
    • 33746883839 scopus 로고    scopus 로고
    • Expression of disease-causing lamin A mutants impairs the formation of DNA repair foci
    • COI: 1:CAS:528:DC%2BD28XnvF2iur8%3D, PID: 16772334
    • Manju K, Muralikrishna B, Parnaik VK (2006) Expression of disease-causing lamin A mutants impairs the formation of DNA repair foci. J Cell Sci 119(13):2704–2714. doi:10.1242/jcs.03009
    • (2006) J Cell Sci , vol.119 , Issue.13 , pp. 2704-2714
    • Manju, K.1    Muralikrishna, B.2    Parnaik, V.K.3
  • 108
    • 0024006572 scopus 로고
    • Increased secretion of fibronectin and collagen by progeria (Hutchinson–Gilford) fibroblasts
    • COI: 1:STN:280:DyaL1c3otlOmtA%3D%3D, PID: 3396601
    • Maquart FX, Bellon G, Gillery P, Borel JP, Labeille B, Risbourg B, Denoeux JP (1988) Increased secretion of fibronectin and collagen by progeria (Hutchinson–Gilford) fibroblasts. Eur J Pediatr 147(4):442
    • (1988) Eur J Pediatr , vol.147 , Issue.4 , pp. 442
    • Maquart, F.X.1    Bellon, G.2    Gillery, P.3    Borel, J.P.4    Labeille, B.5    Risbourg, B.6    Denoeux, J.P.7
  • 109
    • 33947715599 scopus 로고    scopus 로고
    • Barrier-to-autointegration factor—a BAFfling little protein
    • COI: 1:CAS:528:DC%2BD2sXjvFGisbk%3D, PID: 17320395
    • Margalit A, Brachner A, Gotzmann J, Foisner R, Gruenbaum Y (2007) Barrier-to-autointegration factor—a BAFfling little protein. Trends Cell Biol 17(4):202–208
    • (2007) Trends Cell Biol , vol.17 , Issue.4 , pp. 202-208
    • Margalit, A.1    Brachner, A.2    Gotzmann, J.3    Foisner, R.4    Gruenbaum, Y.5
  • 110
  • 111
    • 1842854443 scopus 로고    scopus 로고
    • Lamin A/C binding protein LAP2α is required for nuclear anchorage of retinoblastoma protein
    • COI: 1:CAS:528:DC%2BD38XpsFOqs78%3D, PID: 12475961
    • Markiewicz E, Dechat T, Foisner R, Quinlan RA, Hutchison CJ (2002) Lamin A/C binding protein LAP2α is required for nuclear anchorage of retinoblastoma protein. Mol Biol Cell 13(12):4401–4413. doi:10.1091/mbc.E02-07-0450
    • (2002) Mol Biol Cell , vol.13 , Issue.12 , pp. 4401-4413
    • Markiewicz, E.1    Dechat, T.2    Foisner, R.3    Quinlan, R.A.4    Hutchison, C.J.5
  • 112
    • 41649101975 scopus 로고    scopus 로고
    • The mutant form of Lamin A that causes Hutchinson–Gilford progeria is a biomarker of cellular aging in human skin
    • PID: 18060063
    • McClintock D, Ratner D, Lokuge M, Owens DM, Gordon LB, Collins FS, Djabali K (2007) The mutant form of Lamin A that causes Hutchinson–Gilford progeria is a biomarker of cellular aging in human skin. PLoS ONE. doi:10.1371/journal.pone.0001269
    • (2007) PLoS ONE
    • McClintock, D.1    Ratner, D.2    Lokuge, M.3    Owens, D.M.4    Gordon, L.B.5    Collins, F.S.6    Djabali, K.7
  • 113
    • 84873349707 scopus 로고    scopus 로고
    • Correlated alterations in genome organization, histone methylation, and DNA-lamin A/C interactions in Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BC3sXitVemt7o%3D, PID: 23152449
    • McCord RP, Nazario-Toole A, Zhang H, Chines PS, Zhan Y, Erdos MR, Collins FS, Dekker J, Cao K (2013) Correlated alterations in genome organization, histone methylation, and DNA-lamin A/C interactions in Hutchinson–Gilford progeria syndrome. Genome Res 23(2):260–269. doi:10.1101/gr.138032.112
    • (2013) Genome Res , vol.23 , Issue.2 , pp. 260-269
    • McCord, R.P.1    Nazario-Toole, A.2    Zhang, H.3    Chines, P.S.4    Zhan, Y.5    Erdos, M.R.6    Collins, F.S.7    Dekker, J.8    Cao, K.9
  • 114
    • 63749130030 scopus 로고    scopus 로고
    • Ovarian failure and dilated cardiomyopathy due to a novel lamin mutation
    • COI: 1:CAS:528:DC%2BD1MXksFensbw%3D, PID: 19283854
    • McPherson E, Turner L, Zador I, Reynolds K, Macgregor D, Giampietro PF (2009) Ovarian failure and dilated cardiomyopathy due to a novel lamin mutation. Am J Med Genet A 149A(4):567–572. doi:10.1002/ajmg.a.32627
    • (2009) Am J Med Genet A , vol.149A , Issue.4 , pp. 567-572
    • McPherson, E.1    Turner, L.2    Zador, I.3    Reynolds, K.4    Macgregor, D.5    Giampietro, P.F.6
  • 115
    • 70349636048 scopus 로고    scopus 로고
    • NURD keeps chromatin young
    • COI: 1:CAS:528:DC%2BD1MXht1SqsLfO, PID: 19794502
    • Meshorer E, Gruenbaum Y (2009) NURD keeps chromatin young. Nat Cell Biol 11(10):1176–1177. doi:10.1038/ncb1009-1176
    • (2009) Nat Cell Biol , vol.11 , Issue.10 , pp. 1176-1177
    • Meshorer, E.1    Gruenbaum, Y.2
  • 116
    • 84873375779 scopus 로고    scopus 로고
    • Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence
    • COI: 1:CAS:528:DC%2BC3sXitVemt7s%3D, PID: 23124521
    • Meuleman W, Peric-Hupkes D, Kind J, Beaudry JB, Pagie L, Kellis M, Reinders M, Wessels L, van Steensel B (2013) Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence. Genome Res 23(2):270–280. doi:10.1101/gr.141028.112
    • (2013) Genome Res , vol.23 , Issue.2 , pp. 270-280
    • Meuleman, W.1    Peric-Hupkes, D.2    Kind, J.3    Beaudry, J.B.4    Pagie, L.5    Kellis, M.6    Reinders, M.7    Wessels, L.8    van Steensel, B.9
  • 119
    • 0034638842 scopus 로고    scopus 로고
    • Nuclear lamins A and B1: different pathways of assembly during nuclear envelope formation in living cells
    • COI: 1:CAS:528:DC%2BD3cXovFant7k%3D, PID: 11121432
    • Moir RD, Yoon M, Khuon S, Goldman RD (2000) Nuclear lamins A and B1: different pathways of assembly during nuclear envelope formation in living cells. J Cell Biol 151(6):1155–1168
    • (2000) J Cell Biol , vol.151 , Issue.6 , pp. 1155-1168
    • Moir, R.D.1    Yoon, M.2    Khuon, S.3    Goldman, R.D.4
  • 120
    • 33646088738 scopus 로고    scopus 로고
    • Cell surface expression of intermediate filament proteins vimentin and lamin B1 in human neutrophil spontaneous apoptosis
    • COI: 1:CAS:528:DC%2BD28XisFKnu74%3D, PID: 16365157
    • Moisan E, Girard D (2006) Cell surface expression of intermediate filament proteins vimentin and lamin B1 in human neutrophil spontaneous apoptosis. J Leukoc Biol 79(3):489–498. doi:10.1189/jlb.0405190
    • (2006) J Leukoc Biol , vol.79 , Issue.3 , pp. 489-498
    • Moisan, E.1    Girard, D.2
  • 121
    • 58849099548 scopus 로고    scopus 로고
    • Atypical generalized lipoatrophy and severe insulin resistance due to a heterozygous LMNA p.T101 mutation
    • PID: 19169477
    • Mory PB, Crispim F, Kasamatsu T, Gabbay MAL, Dib SA, Moises RS (2008) Atypical generalized lipoatrophy and severe insulin resistance due to a heterozygous LMNA p.T101 mutation. Arq Bras Endocrinol Metabol 52(8):1252–1256
    • (2008) Arq Bras Endocrinol Metabol , vol.52 , Issue.8 , pp. 1252-1256
    • Mory, P.B.1    Crispim, F.2    Kasamatsu, T.3    Gabbay, M.A.L.4    Dib, S.A.5    Moises, R.S.6
  • 122
    • 0037673940 scopus 로고    scopus 로고
    • A progeroid syndrome in mice is caused by defects in A-type lamins
    • COI: 1:CAS:528:DC%2BD3sXjs1ynurc%3D, PID: 12748643
    • Mounkes LC, Kozlov S, Hernandez L, Sullivan T, Stewart CL (2003) A progeroid syndrome in mice is caused by defects in A-type lamins. Nature 423(6937):298–301. doi:10.1038/nature01631
    • (2003) Nature , vol.423 , Issue.6937 , pp. 298-301
    • Mounkes, L.C.1    Kozlov, S.2    Hernandez, L.3    Sullivan, T.4    Stewart, C.L.5
  • 123
    • 77956578554 scopus 로고    scopus 로고
    • Signaling defects and the nuclear envelope in progeria
    • COI: 1:CAS:528:DC%2BC3cXhtFGht7zE, PID: 20833355
    • Muchir A, Worman HJ (2010) Signaling defects and the nuclear envelope in progeria. Dev Cell 19(3):355–356. doi:10.1016/j.devcel.2010.08.019
    • (2010) Dev Cell , vol.19 , Issue.3 , pp. 355-356
    • Muchir, A.1    Worman, H.J.2
  • 124
    • 81855198154 scopus 로고    scopus 로고
    • DNA-damage accumulation and replicative arrest in Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BC3MXhsFaiur3K, PID: 22103522
    • Musich PR, Zou Y (2011) DNA-damage accumulation and replicative arrest in Hutchinson–Gilford progeria syndrome. Biochem Soc Trans 39:1764–1769. doi:10.1042/Bst20110687
    • (2011) Biochem Soc Trans , vol.39 , pp. 1764-1769
    • Musich, P.R.1    Zou, Y.2
  • 127
    • 33748993124 scopus 로고    scopus 로고
    • Molecular bases of progeroid syndromes
    • COI: 1:CAS:528:DC%2BD28XpvFGlsLY%3D, PID: 16987878
    • Navarro CL, Cau P, Levy N (2006) Molecular bases of progeroid syndromes. Hum Mol Genet 15(2):R151–R161. doi:10.1093/hmg/ddl214
    • (2006) Hum Mol Genet , vol.15 , Issue.2 , pp. R151-R161
    • Navarro, C.L.1    Cau, P.2    Levy, N.3
  • 128
    • 84866183226 scopus 로고    scopus 로고
    • Ageing as a risk factor for disease
    • COI: 1:CAS:528:DC%2BC38XhtlCjtLbK, PID: 22975005
    • Niccoli T, Partridge L (2012) Ageing as a risk factor for disease. Curr Biol 22(17):R741–R752. doi:10.1016/j.cub.2012.07.024
    • (2012) Curr Biol , vol.22 , Issue.17 , pp. R741-R752
    • Niccoli, T.1    Partridge, L.2
  • 129
    • 41449094144 scopus 로고    scopus 로고
    • Evidence that proteasome-dependent degradation of the retinoblastoma protein in cells lacking A-type lamins occurs independently of gankyrin and MDM2
    • PID: 17896003
    • Nitta RT, Smith CL, Kennedy BK (2007) Evidence that proteasome-dependent degradation of the retinoblastoma protein in cells lacking A-type lamins occurs independently of gankyrin and MDM2. PLoS ONE. doi:10.1371/journal.pone.0000963
    • (2007) PLoS ONE
    • Nitta, R.T.1    Smith, C.L.2    Kennedy, B.K.3
  • 131
    • 84922154914 scopus 로고    scopus 로고
    • Lamins at the crossroads of mechanosignaling
    • COI: 1:CAS:528:DC%2BC2MXjsVyitbk%3D, PID: 25644599
    • Osmanagic-Myers S, Dechat T, Foisner R (2015) Lamins at the crossroads of mechanosignaling. Genes Dev 29(3):225–237. doi:10.1101/gad.255968.114
    • (2015) Genes Dev , vol.29 , Issue.3 , pp. 225-237
    • Osmanagic-Myers, S.1    Dechat, T.2    Foisner, R.3
  • 133
  • 134
    • 70349645266 scopus 로고    scopus 로고
    • Ageing-related chromatin defects through loss of the NURD complex
    • COI: 1:CAS:528:DC%2BD1MXht1SqsLnP, PID: 19734887
    • Pegoraro G, Kubben N, Wickert U, Gohler H, Hoffmann K, Misteli T (2009) Ageing-related chromatin defects through loss of the NURD complex. Nat Cell Biol 11(10):1261–1267. doi:10.1038/ncb1971
    • (2009) Nat Cell Biol , vol.11 , Issue.10 , pp. 1261-1267
    • Pegoraro, G.1    Kubben, N.2    Wickert, U.3    Gohler, H.4    Hoffmann, K.5    Misteli, T.6
  • 136
    • 84945143871 scopus 로고    scopus 로고
    • All-trans retinoic acid and rapamycin normalize Hutchinson–Gilford progeria fibroblast phenotype
    • PID: 26359359
    • Pellegrini C, Columbaro M, Capanni C, D’Apice MR, Cavallo C, Murdocca M, Lattanzi G, Squarzoni S (2015) All-trans retinoic acid and rapamycin normalize Hutchinson–Gilford progeria fibroblast phenotype. Oncotarget 6(30):29914–29928. doi:10.18632/oncotarget.4939
    • (2015) Oncotarget , vol.6 , Issue.30 , pp. 29914-29928
    • Pellegrini, C.1    Columbaro, M.2    Capanni, C.3    D’Apice, M.R.4    Cavallo, C.5    Murdocca, M.6    Lattanzi, G.7    Squarzoni, S.8
  • 138
    • 45449092970 scopus 로고    scopus 로고
    • HGPS and related premature aging disorders: from genomic identification to the first therapeutic approaches
    • COI: 1:CAS:528:DC%2BD1cXns1Shurs%3D, PID: 18513784
    • Pereira S, Bourgeois P, Navarro C, Esteves-Vieira V, Cau P, De Sandre-Giovannoli A, Levy N (2008) HGPS and related premature aging disorders: from genomic identification to the first therapeutic approaches. Mech Ageing Dev 129(7–8):449–459. doi:10.1016/j.mad.2008.04.003
    • (2008) Mech Ageing Dev , vol.129 , Issue.7-8 , pp. 449-459
    • Pereira, S.1    Bourgeois, P.2    Navarro, C.3    Esteves-Vieira, V.4    Cau, P.5    De Sandre-Giovannoli, A.6    Levy, N.7
  • 140
    • 55549113990 scopus 로고    scopus 로고
    • Nuclear mechanotransduction: response of the lamina to extracellular stress with implications in aging
    • PID: 18945430
    • Philip JT, Dahl KN (2008) Nuclear mechanotransduction: response of the lamina to extracellular stress with implications in aging. J Biomech 41(15):3164–3170. doi:10.1016/j.jbiomech.2008.08.024
    • (2008) J Biomech , vol.41 , Issue.15 , pp. 3164-3170
    • Philip, J.T.1    Dahl, K.N.2
  • 141
    • 84880513010 scopus 로고    scopus 로고
    • Hutchinson–Gilford progeria syndrome through the lens of transcription
    • COI: 1:CAS:528:DC%2BC3sXhtlCku7jE, PID: 23496208
    • Prokocimer M, Barkan R, Gruenbaum Y (2013) Hutchinson–Gilford progeria syndrome through the lens of transcription. Aging Cell 12(4):533–543. doi:10.1111/acel.12070
    • (2013) Aging Cell , vol.12 , Issue.4 , pp. 533-543
    • Prokocimer, M.1    Barkan, R.2    Gruenbaum, Y.3
  • 143
    • 80053150156 scopus 로고    scopus 로고
    • The accumulation of un-repairable DNA damage in laminopathy progeria fibroblasts is caused by ROS generation and is prevented by treatment with N-acetyl cysteine
    • COI: 1:CAS:528:DC%2BC3MXht1ajtbnN, PID: 21807766
    • Richards SA, Muter J, Ritchie P, Lattanzi G, Hutchison CJ (2011) The accumulation of un-repairable DNA damage in laminopathy progeria fibroblasts is caused by ROS generation and is prevented by treatment with N-acetyl cysteine. Hum Mol Genet 20(20):3997–4004. doi:10.1093/hmg/ddr327
    • (2011) Hum Mol Genet , vol.20 , Issue.20 , pp. 3997-4004
    • Richards, S.A.1    Muter, J.2    Ritchie, P.3    Lattanzi, G.4    Hutchison, C.J.5
  • 144
    • 84956644335 scopus 로고    scopus 로고
    • Prepatterning of differentiation-driven nuclear lamin A/C-associated chromatin domains by GlcNAcylated histone H2B
    • PID: 26359231
    • Ronningen T, Shah A, Oldenburg AR, Vekterud K, Delbarre E, Moskaug JO, Collas P (2015) Prepatterning of differentiation-driven nuclear lamin A/C-associated chromatin domains by GlcNAcylated histone H2B. Genome Res 25(12):1825–1835. doi:10.1101/gr.193748.115
    • (2015) Genome Res , vol.25 , Issue.12 , pp. 1825-1835
    • Ronningen, T.1    Shah, A.2    Oldenburg, A.R.3    Vekterud, K.4    Delbarre, E.5    Moskaug, J.O.6    Collas, P.7
  • 145
    • 81155125086 scopus 로고    scopus 로고
    • Stem cell depletion in Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BC3MXhs1Sqt7rJ, PID: 21902803
    • Rosengardten Y, McKenna T, Grochova D, Eriksson M (2011) Stem cell depletion in Hutchinson–Gilford progeria syndrome. Aging Cell 10(6):1011–1020. doi:10.1111/j.1474-9726.2011.00743.x
    • (2011) Aging Cell , vol.10 , Issue.6 , pp. 1011-1020
    • Rosengardten, Y.1    McKenna, T.2    Grochova, D.3    Eriksson, M.4
  • 146
    • 33748760066 scopus 로고    scopus 로고
    • Farnesylated lamins, progeroid syndromes and farnesyl transferase inhibitors
    • COI: 1:CAS:528:DC%2BD28XhtVart7zF, PID: 16899817
    • Rusinol AE, Sinensky MS (2006) Farnesylated lamins, progeroid syndromes and farnesyl transferase inhibitors. J Cell Sci 119(Pt 16):3265–3272. doi:10.1242/jcs.03156
    • (2006) J Cell Sci , vol.119 , pp. 3265-3272
    • Rusinol, A.E.1    Sinensky, M.S.2
  • 147
    • 43149117884 scopus 로고    scopus 로고
    • Targeted transgenic expression of the mutation causing Hutchinson–Gilford progeria syndrome leads to proliferative and degenerative epidermal disease
    • COI: 1:CAS:528:DC%2BD1cXlsF2nsbw%3D, PID: 18334552
    • Sagelius H, Rosengardten Y, Hanif M, Erdos MR, Rozell B, Collins FS, Eriksson M (2008) Targeted transgenic expression of the mutation causing Hutchinson–Gilford progeria syndrome leads to proliferative and degenerative epidermal disease. J Cell Sci 121(Pt 7):969–978. doi:10.1242/jcs.022913
    • (2008) J Cell Sci , vol.121 , pp. 969-978
    • Sagelius, H.1    Rosengardten, Y.2    Hanif, M.3    Erdos, M.R.4    Rozell, B.5    Collins, F.S.6    Eriksson, M.7
  • 148
    • 77950265945 scopus 로고    scopus 로고
    • Linking functional decline of telomeres, mitochondria and stem cells during ageing
    • COI: 1:CAS:528:DC%2BC3cXjvVGlsbo%3D, PID: 20336134
    • Sahin E, Depinho RA (2010) Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature 464(7288):520–528. doi:10.1038/nature08982
    • (2010) Nature , vol.464 , Issue.7288 , pp. 520-528
    • Sahin, E.1    Depinho, R.A.2
  • 149
    • 17644373758 scopus 로고    scopus 로고
    • (R)eversal of the cellular phenotype in the premature aging disease Hutchinson–Gilford progeria syndrome
    • COI: 1:CAS:528:DC%2BD2MXivFCgsr0%3D, PID: 15750600
    • Scaffidi P, Misteli T (2005) (R)eversal of the cellular phenotype in the premature aging disease Hutchinson–Gilford progeria syndrome. Nat Med 11(4):440–445. doi:10.1038/nm1204
    • (2005) Nat Med , vol.11 , Issue.4 , pp. 440-445
    • Scaffidi, P.1    Misteli, T.2
  • 150
    • 33646745137 scopus 로고    scopus 로고
    • Lamin A-dependent nuclear defects in human aging
    • COI: 1:CAS:528:DC%2BD28Xks1Wqur0%3D, PID: 16645051
    • Scaffidi P, Misteli T (2006) Lamin A-dependent nuclear defects in human aging. Science 312(5776):1059–1063. doi:10.1126/science.1127168
    • (2006) Science , vol.312 , Issue.5776 , pp. 1059-1063
    • Scaffidi, P.1    Misteli, T.2
  • 151
    • 43149124203 scopus 로고    scopus 로고
    • Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing
    • COI: 1:CAS:528:DC%2BD1cXktVGqtrw%3D, PID: 18311132
    • Scaffidi P, Misteli T (2008) Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat Cell Biol 10(4):452–459. doi:10.1038/ncb1708
    • (2008) Nat Cell Biol , vol.10 , Issue.4 , pp. 452-459
    • Scaffidi, P.1    Misteli, T.2
  • 152
    • 84866939080 scopus 로고    scopus 로고
    • Expression of the Hutchinson–Gilford progeria mutation during osteoblast development results in loss of osteocytes, irregular mineralization, and poor biomechanical properties
    • COI: 1:CAS:528:DC%2BC38XhsVarsLjN, PID: 22893709
    • Schmidt E, Nilsson O, Koskela A, Tuukkanen J, Ohlsson C, Rozell B, Eriksson M (2012) Expression of the Hutchinson–Gilford progeria mutation during osteoblast development results in loss of osteocytes, irregular mineralization, and poor biomechanical properties. J Biol Chem 287(40):33512–33522. doi:10.1074/jbc.M112.366450
    • (2012) J Biol Chem , vol.287 , Issue.40 , pp. 33512-33522
    • Schmidt, E.1    Nilsson, O.2    Koskela, A.3    Tuukkanen, J.4    Ohlsson, C.5    Rozell, B.6    Eriksson, M.7
  • 156
    • 84899407577 scopus 로고    scopus 로고
    • Shear stress-induced mechanotransduction protein deregulation and vasculopathy in a mouse model of progeria
    • PID: 24661531
    • Song M, San H, Anderson SA, Cannon RO 3rd, Orlic D (2014) Shear stress-induced mechanotransduction protein deregulation and vasculopathy in a mouse model of progeria. Stem Cell Res Ther 5(2):41. doi:10.1186/scrt429
    • (2014) Stem Cell Res Ther , vol.5 , Issue.2 , pp. 41
    • Song, M.1    San, H.2    Anderson, S.A.3    Cannon, R.O.4    Orlic, D.5
  • 157
    • 0032943657 scopus 로고    scopus 로고
    • Histological and ultrastructural features of atherosclerosis in progeria
    • COI: 1:STN:280:DC%2BD3c7osFehsw%3D%3D, PID: 10722246
    • Stehbens WE, Wakefield SJ, Gilbert-Barness E, Olson RE, Ackerman J (1999) Histological and ultrastructural features of atherosclerosis in progeria. Cardiovasc Pathol 8(1):29–39
    • (1999) Cardiovasc Pathol , vol.8 , Issue.1 , pp. 29-39
    • Stehbens, W.E.1    Wakefield, S.J.2    Gilbert-Barness, E.3    Olson, R.E.4    Ackerman, J.5
  • 158
    • 84940399428 scopus 로고    scopus 로고
    • Transgene silencing of the Hutchinson–Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does not show overall beneficial effects
    • COI: 1:CAS:528:DC%2BC28Xmslaiug%3D%3D, PID: 25877214
    • Strandgren C, Nasser HA, McKenna T, Koskela A, Tuukkanen J, Ohlsson C, Rozell B, Eriksson M (2015) Transgene silencing of the Hutchinson–Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does not show overall beneficial effects. FASEB J 29(8):3193–3205. doi:10.1096/fj.14-269217
    • (2015) FASEB J , vol.29 , Issue.8 , pp. 3193-3205
    • Strandgren, C.1    Nasser, H.A.2    McKenna, T.3    Koskela, A.4    Tuukkanen, J.5    Ohlsson, C.6    Rozell, B.7    Eriksson, M.8
  • 159
    • 84895763410 scopus 로고    scopus 로고
    • Label-free mass spectrometry exploits dozens of detected peptides to quantify lamins in wildtype and knockdown cells
    • PID: 24448480
    • Swift J, Harada T, Buxboim A, Shin JW, Tang HY, Speicher DW, Discher DE (2013) Label-free mass spectrometry exploits dozens of detected peptides to quantify lamins in wildtype and knockdown cells. Nucleus 4(6):450–459. doi:10.4161/nucl.27413
    • (2013) Nucleus , vol.4 , Issue.6 , pp. 450-459
    • Swift, J.1    Harada, T.2    Buxboim, A.3    Shin, J.W.4    Tang, H.Y.5    Speicher, D.W.6    Discher, D.E.7
  • 160
    • 84934440388 scopus 로고    scopus 로고
    • LC3 and autophagy
    • COI: 1:CAS:528:DC%2BD1cXmsV2lsrs%3D, PID: 18425443
    • Tanida I, Ueno T, Kominami E (2008) LC3 and autophagy. Methods Mol Biol 445:77–88. doi:10.1007/978-1-59745-157-4_4
    • (2008) Methods Mol Biol , vol.445 , pp. 77-88
    • Tanida, I.1    Ueno, T.2    Kominami, E.3
  • 162
    • 24644459728 scopus 로고    scopus 로고
    • Blocking protein farnesyltransferase improves nuclear shape in fibroblasts from humans with progeroid syndromes
    • COI: 1:CAS:528:DC%2BD2MXhtVWktb7L, PID: 16129834
    • Toth JI, Yang SH, Qiao X, Beigneux AP, Gelb MH, Moulson CL, Miner JH, Young SG, Fong LG (2005) Blocking protein farnesyltransferase improves nuclear shape in fibroblasts from humans with progeroid syndromes. Proc Natl Acad Sci USA 102(36):12873–12878. doi:10.1073/pnas.0505767102
    • (2005) Proc Natl Acad Sci USA , vol.102 , Issue.36 , pp. 12873-12878
    • Toth, J.I.1    Yang, S.H.2    Qiao, X.3    Beigneux, A.P.4    Gelb, M.H.5    Moulson, C.L.6    Miner, J.H.7    Young, S.G.8    Fong, L.G.9
  • 167
    • 34248391873 scopus 로고    scopus 로고
    • The nuclear envelope, a key structure in cellular integrity and gene expression
    • COI: 1:CAS:528:DC%2BD2sXltFejsrY%3D, PID: 17504143
    • Verstraeten VLRM, Broers JLV, Ramaekers FCS, van Steensel MAM (2007) The nuclear envelope, a key structure in cellular integrity and gene expression. Curr Med Chem 14(11):1231–1248
    • (2007) Curr Med Chem , vol.14 , Issue.11 , pp. 1231-1248
    • Verstraeten, V.L.R.M.1    Broers, J.L.V.2    Ramaekers, F.C.S.3    van Steensel, M.A.M.4
  • 168
    • 43449096922 scopus 로고    scopus 로고
    • Increased mechanosensitivity and nuclear stiffness in Hutchinson–Gilford progeria cells: effects of farnesyltransferase inhibitors
    • COI: 1:CAS:528:DC%2BD1cXntFWnt74%3D, PID: 18331619
    • Verstraeten VL, Ji JY, Cummings KS, Lee RT, Lammerding J (2008) Increased mechanosensitivity and nuclear stiffness in Hutchinson–Gilford progeria cells: effects of farnesyltransferase inhibitors. Aging Cell 7(3):383–393. doi:10.1111/j.1474-9726.2008.00382.x
    • (2008) Aging Cell , vol.7 , Issue.3 , pp. 383-393
    • Verstraeten, V.L.1    Ji, J.Y.2    Cummings, K.S.3    Lee, R.T.4    Lammerding, J.5
  • 169
    • 84943572682 scopus 로고    scopus 로고
    • Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2α (LAP2α) through expression of extracellular matrix proteins
    • COI: 1:CAS:528:DC%2BC2MXhslCku77P, PID: 26443848
    • Vidak S, Kubben N, Dechat T, Foisner R (2015) Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2α (LAP2α) through expression of extracellular matrix proteins. Genes Dev 29(19):2022–2036. doi:10.1101/gad.263939.115
    • (2015) Genes Dev , vol.29 , Issue.19 , pp. 2022-2036
    • Vidak, S.1    Kubben, N.2    Dechat, T.3    Foisner, R.4
  • 170
    • 84964846245 scopus 로고    scopus 로고
    • Naive adult stem cells from patients with Hutchinson–Gilford progeria syndrome express low levels of progerin in vivo
    • COI: 1:CAS:528:DC%2BC38Xpt1Wqurs%3D, PID: 23213444
    • Wenzel V, Roedl D, Gabriel D, Gordon LB, Herlyn M, Schneider R, Ring J, Djabali K (2012) Naive adult stem cells from patients with Hutchinson–Gilford progeria syndrome express low levels of progerin in vivo. Biol Open 1(6):516–526. doi:10.1242/bio.20121149
    • (2012) Biol Open , vol.1 , Issue.6 , pp. 516-526
    • Wenzel, V.1    Roedl, D.2    Gabriel, D.3    Gordon, L.B.4    Herlyn, M.5    Schneider, R.6    Ring, J.7    Djabali, K.8
  • 171
    • 77956083200 scopus 로고    scopus 로고
    • Lamin-binding proteins
    • PID: 20452940
    • Wilson KL, Foisner R (2010) Lamin-binding proteins. Cold Spring Harb Perspect Biol 2(4):a000554. doi:10.1101/cshperspect.a000554
    • (2010) Cold Spring Harb Perspect Biol , vol.2 , Issue.4 , pp. a000554
    • Wilson, K.L.1    Foisner, R.2
  • 172
    • 4544270426 scopus 로고    scopus 로고
    • Sirtuin activators mimic caloric restriction and delay ageing in metazoans
    • COI: 1:CAS:528:DC%2BD2cXntFCks7s%3D, (vol 430, p 686, 2004)
    • Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, Tatar M, Sinclair D (2004) Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 431(7004):107. doi:10.1038/nature02941(vol 430, p 686, 2004)
    • (2004) Nature , vol.431 , Issue.7004 , pp. 107
    • Wood, J.G.1    Rogina, B.2    Lavu, S.3    Howitz, K.4    Helfand, S.L.5    Tatar, M.6    Sinclair, D.7
  • 173
    • 82755161872 scopus 로고    scopus 로고
    • Nuclear lamins and laminopathies
    • COI: 1:CAS:528:DC%2BC3MXhsFCqsbrJ, PID: 21953297
    • Worman HJ (2012) Nuclear lamins and laminopathies. J Pathol 226(2):316–325. doi:10.1002/path.2999
    • (2012) J Pathol , vol.226 , Issue.2 , pp. 316-325
    • Worman, H.J.1
  • 174
    • 84935011432 scopus 로고    scopus 로고
    • Nuclear membrane diversity: underlying tissue-specific pathologies in disease?
    • COI: 1:CAS:528:DC%2BC2MXhtVyiurfM, PID: 26115475
    • Worman HJ, Schirmer EC (2015) Nuclear membrane diversity: underlying tissue-specific pathologies in disease? Curr Opin Cell Biol 34:101–112. doi:10.1016/j.ceb.2015.06.003
    • (2015) Curr Opin Cell Biol , vol.34 , pp. 101-112
    • Worman, H.J.1    Schirmer, E.C.2
  • 175
    • 22544440839 scopus 로고    scopus 로고
    • Blocking protein farnesyltransferase improves nuclear blebbing in mouse fibroblasts with a targeted Hutchinson-Gilford progeria syndrome mutation
    • COI: 1:CAS:528:DC%2BD2MXmvVeks78%3D, PID: 16014412
    • Yang SH, Bergo MO, Toth JI, Qiao X, Hu Y, Sandoval S, Meta M, Bendale P, Gelb MH, Young SG, Fong LG (2005) Blocking protein farnesyltransferase improves nuclear blebbing in mouse fibroblasts with a targeted Hutchinson-Gilford progeria syndrome mutation. Proc Natl Acad Sci USA 102(29):10291–10296. doi:10.1073/pnas.0504641102
    • (2005) Proc Natl Acad Sci USA , vol.102 , Issue.29 , pp. 10291-10296
    • Yang, S.H.1    Bergo, M.O.2    Toth, J.I.3    Qiao, X.4    Hu, Y.5    Sandoval, S.6    Meta, M.7    Bendale, P.8    Gelb, M.H.9    Young, S.G.10    Fong, L.G.11
  • 176
    • 55849129996 scopus 로고    scopus 로고
    • Progerin elicits disease phenotypes of progeria in mice whether or not it is farnesylated
    • COI: 1:CAS:528:DC%2BD1cXht1Chsr3K, PID: 18769635
    • Yang SH, Andres DA, Spielmann HP, Young SG, Fong LG (2008) Progerin elicits disease phenotypes of progeria in mice whether or not it is farnesylated. J Clin Invest 118(10):3291–3300. doi:10.1172/JCI35876
    • (2008) J Clin Invest , vol.118 , Issue.10 , pp. 3291-3300
    • Yang, S.H.1    Andres, D.A.2    Spielmann, H.P.3    Young, S.G.4    Fong, L.G.5
  • 177
    • 79960566670 scopus 로고    scopus 로고
    • Differential expression of extracellular matrix proteins in senescent and young human fibroblasts: a comparative proteomics and microarray study
    • COI: 1:CAS:528:DC%2BC3MXptVyktLc%3D, PID: 21573704
    • Yang KE, Kwon J, Rhim JH, Choi JS, Kim SI, Lee SH, Park J, Jang IS (2011a) Differential expression of extracellular matrix proteins in senescent and young human fibroblasts: a comparative proteomics and microarray study. Mol Cells 32(1):99–106. doi:10.1007/s10059-011-0064-0
    • (2011) Mol Cells , vol.32 , Issue.1 , pp. 99-106
    • Yang, K.E.1    Kwon, J.2    Rhim, J.H.3    Choi, J.S.4    Kim, S.I.5    Lee, S.H.6    Park, J.7    Jang, I.S.8
  • 178
    • 78651095782 scopus 로고    scopus 로고
    • Absence of progeria-like disease phenotypes in knock-in mice expressing a non-farnesylated version of progerin
    • COI: 1:CAS:528:DC%2BC3MXhslKnt78%3D, PID: 21088111
    • Yang SH, Chang SY, Ren S, Wang Y, Andres DA, Spielmann HP, Fong LG, Young SG (2011b) Absence of progeria-like disease phenotypes in knock-in mice expressing a non-farnesylated version of progerin. Hum Mol Genet 20(3):436–444. doi:10.1093/hmg/ddq490
    • (2011) Hum Mol Genet , vol.20 , Issue.3 , pp. 436-444
    • Yang, S.H.1    Chang, S.Y.2    Ren, S.3    Wang, Y.4    Andres, D.A.5    Spielmann, H.P.6    Fong, L.G.7    Young, S.G.8
  • 179
    • 30844434561 scopus 로고    scopus 로고
    • Prelamin A, Zmpste24, misshapen cell nuclei, and progeria–new evidence suggesting that protein farnesylation could be important for disease pathogenesis
    • COI: 1:CAS:528:DC%2BD2MXhtlWmsLbI, PID: 16207929
    • Young SG, Fong LG, Michaelis S (2005) Prelamin A, Zmpste24, misshapen cell nuclei, and progeria–new evidence suggesting that protein farnesylation could be important for disease pathogenesis. J Lipid Res 46(12):2531–2558. doi:10.1194/jlr.R500011-JLR200
    • (2005) J Lipid Res , vol.46 , Issue.12 , pp. 2531-2558
    • Young, S.G.1    Fong, L.G.2    Michaelis, S.3
  • 180
    • 78650995671 scopus 로고    scopus 로고
    • A human iPSC model of Hutchinson–Gilford progeria reveals vascular smooth muscle and mesenchymal stem cell defects
    • COI: 1:CAS:528:DC%2BC3MXjvF2msw%3D%3D, PID: 21185252
    • Zhang J, Lian Q, Zhu G, Zhou F, Sui L, Tan C, Mutalif RA, Navasankari R, Zhang Y, Tse HF, Stewart CL, Colman A (2011) A human iPSC model of Hutchinson–Gilford progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 8(1):31–45. doi:10.1016/j.stem.2010.12.002
    • (2011) Cell Stem Cell , vol.8 , Issue.1 , pp. 31-45
    • Zhang, J.1    Lian, Q.2    Zhu, G.3    Zhou, F.4    Sui, L.5    Tan, C.6    Mutalif, R.A.7    Navasankari, R.8    Zhang, Y.9    Tse, H.F.10    Stewart, C.L.11    Colman, A.12
  • 181
    • 84880071424 scopus 로고    scopus 로고
    • High mobility group protein N5 (HMGN5) and lamina-associated polypeptide 2α (LAP2α) interact and reciprocally affect their genome-wide chromatin organization
    • COI: 1:CAS:528:DC%2BC3sXpvVGntLY%3D, PID: 23673662
    • Zhang S, Schones DE, Malicet C, Rochman M, Zhou M, Foisner R, Bustin M (2013) High mobility group protein N5 (HMGN5) and lamina-associated polypeptide 2α (LAP2α) interact and reciprocally affect their genome-wide chromatin organization. J Biol Chem 288(25):18104–18109. doi:10.1074/jbc.C113.469544
    • (2013) J Biol Chem , vol.288 , Issue.25 , pp. 18104-18109
    • Zhang, S.1    Schones, D.E.2    Malicet, C.3    Rochman, M.4    Zhou, M.5    Foisner, R.6    Bustin, M.7


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.