Skip to main content

RB and Lamins in Cell Cycle Regulation and Aging

  • Chapter
  • First Online:
Cancer Biology and the Nuclear Envelope

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 773))

Abstract

While speculation has centered on a role for nuclear lamins in tumor progression for many years, most of the diseases that have been linked to lamin mutation are dystrophic in nature, often limiting the proliferation potential of affected cells in vivo and in vitro. Nevertheless, these lamin mutations, particularly in the LMNA gene that encodes A-type lamins, have provided an interesting tool set to understand functions of nuclear intermediate filament proteins in cell cycle progress and various means of exit, including quiescence, senescence, and differentiation down various lineages. The picture that has emerged is complex with lamins controlling the activity of key cell cycle factors such as the retinoblastoma protein (RB) and interacting with several important signal transduction pathways. Here we describe the current state of knowledge and speculate that lamins may be intimately involved in the regulation of cell proliferation, acting at the interface between cancer and aging.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

ATR:

Ataxia telangiectasia and Rad3-related protein

ADLD:

Autosomal dominant leukodystrophy

CDK:

Cyclin-dependent kinase

DCM1A:

Dilated cardiomyopathy type 1A

ERK:

Extracellular signal-regulated kinases

HGPS:

Hutchinson–Gilford Progeria syndrome

LAP2α:

Lamina-associated polypeptide 2α

MDM2:

Mouse double minute 2 homolog

ROS:

Reactive oxygen species

RB:

Retinoblastoma protein

SASP:

Senescence-associated secretory phenotype

SIRT1:

Silent mating-type information regulation 2 homolog 1

VHL:

von Hippel–Lindau gene

References

  1. Gerace L, Blum A, Blobel G (1978) Immunocytochemical localization of the major polypeptides of the nuclear pore complex-lamina fraction. Interphase and mitotic distribution. J Cell Biol 79(2 Pt 1):546–566

    Article  PubMed  CAS  Google Scholar 

  2. Schreiber KH, Kennedy BK (2013) When lamins go bad: nuclear structure and disease. Cell 152(6):1365–1375

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  3. Gerace L, Blobel G (1980) the nuclear envelope lamina is reversibly depolymerized during mitosis. Cell 19:277–287

    Article  PubMed  CAS  Google Scholar 

  4. Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N, Nagashima K, Stewart CL, Burke B (1999) Loss of A-type lamin expression compromises nuclear envelope integrity leading to muscular dystrophy. J Cell Biol 147:913–920

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  5. Cao H, Hegele RA (2003) LMNA is mutated in Hutchinson-Gilford progeria (MIM 176670) but not in Wiedemann-Rautenstrauch progeroid syndrome (MIM 264090). J Hum Genet 48:271–274

    Article  PubMed  CAS  Google Scholar 

  6. De Sandre-Giovannoli A, Bernard R, Cau P, Navarro C, Amiel J, Boccaccio I, Lyonnet S, Stewart CL, Munnich A, Le Merrer M, Levy N (2003) Lamin A truncation in Hutchison-Gilford progeria. Science 300:2055

    Article  PubMed  Google Scholar 

  7. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, Erdos MR, Robbins CM, Moses TY, Berglund P, Dutra A, Pak E, Durkin S, Csoka AB, Boehnke M, Glover TW, Collins FS (2003) Recurrent de novo point mutations in lamin A cause Hutchison-Gilford progeria syndrome. Nature 423:293–298

    Article  PubMed  CAS  Google Scholar 

  8. Burtner CR, Kennedy BK (2010) Progeria syndromes and ageing: what is the connection? Nat Rev Mol Cell Biol 11(8):567–578

    Article  PubMed  CAS  Google Scholar 

  9. Reddy S, Comai L (2012) Lamin A, farnesylation and aging. Exp Cell Res 318(1):1–7

    Article  PubMed  CAS  Google Scholar 

  10. Verdy C, Branka JE, Mekideche N (2011) Quantitative assessment of lactate and progerin production in normal human cutaneous cells during normal ageing: effect of an Alaria esculenta extract. Int J Cosmet Sci 33(5):462–466

    Article  PubMed  CAS  Google Scholar 

  11. 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 2(12):e1269

    Article  PubMed Central  PubMed  Google Scholar 

  12. Scaffidi P, Misteli T (2006) Lamin A-dependent nuclear defects in human aging. Science 312(5776):1059–1063

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  13. Scaffidi P, Misteli T (2008) Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat Cell Biol 10(4):452–459

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  14. Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Goldman AE, Lucas CA, Shumaker DK, Kosak ST, Chandel NS, Goldman RD (2011) The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev 25(24):2579–2593

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  15. Rober RA, Weber K, Osborn M (1989) Differential timing of nuclear lamin A/C expression in the various organs of the mouse embryo and the young animal: a developmental study. Development 105:365–378

    PubMed  CAS  Google Scholar 

  16. Ho JC, Zhou T, Lai WH, Huang Y, Chan YC, Li X, Wong NL, Li Y, Au KW, Guo D, Xu J, Siu CW, Pei D, Tse HF, Esteban MA (2011) Generation of induced pluripotent stem cell lines from 3 distinct laminopathies bearing heterogeneous mutations in lamin A/C. Aging (Albany NY) 3(4):380–390

    CAS  Google Scholar 

  17. 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

    Article  PubMed  CAS  Google Scholar 

  18. Liu GH, Barkho BZ, Ruiz S, Diep D, Qu J, Yang SL, Panopoulos AD, Suzuki K, Kurian L, Walsh C, Thompson J, Boue S, Fung HL, Sancho-Martinez I, Zhang K, Yates J 3rd, Izpisua Belmonte JC (2011) Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature 472(7342):221–225

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  19. Eisenman RN, Tachibana CY, Abrams HD, Hann SR (1985) V-myc- and c-myc-encoded proteins are associated with the nuclear matrix. Mol Cell Biol 5(1):114–126

    PubMed Central  PubMed  CAS  Google Scholar 

  20. Mancini MA, Shan B, Nickerson JA, Penman S, Lee WH (1994) The retinoblastoma gene product is a cell cycle-dependent, nuclear matrix-associated protein. Proc Natl Acad Sci U S A 91(1):418–422

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  21. Ozaki T, Saijo M, Murakami K, Enomoto H, Taya Y, Sakiyama S (1994) Complex formation between Lamin A and the retinoblastoma gene product: identification of the domain on Lamin A required for its interaction. Oncogene 9:2649–2653

    PubMed  CAS  Google Scholar 

  22. Templeton DJ, Park SH, Lanier L, Weinberg RA (1991) Nonfunctional mutants of the retinoblastoma protein are characterized by defects in phosphorylation, viral oncoprotein association, and nuclear tethering. Proc Natl Acad Sci U S A 88:3033–3037

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  23. Mittnacht S, Weinberg RA (1991) G1/S phosphorylation of the retinoblastoma protein is associated with altered affinity for the nuclear compartment. Cell 65:381–393

    Article  PubMed  CAS  Google Scholar 

  24. Redwood AB, Gonzalez-Suarez I, Gonzalo S (2011) Regulating the levels of key factors in cell cycle and DNA repair: new pathways revealed by lamins. Cell Cycle 10(21):3652–3657

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  25. Zink D, Fischer AH, Nickerson JA (2004) Nuclear structure in cancer cells. Nat Rev Cancer 4(9):677–687

    Article  PubMed  CAS  Google Scholar 

  26. Foster CR, Przyborski SA, Wilson RG, Hutchison CJ (2010) Lamins as cancer biomarkers. Biochem Soc Trans 38(Pt 1):297–300

    Article  PubMed  CAS  Google Scholar 

  27. Butin-Israeli V, Adam SA, Goldman AE, Goldman RD (2012) Nuclear lamin functions and disease. Trends Genet 28(9):464–471

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  28. Nikolova V, Leimena C, McMahon AC, Tan JC, Chandar S, Jogia D, Kesteven SH, Michalicek J, Otway R, Verheyen F, Rainer S, Stewart CL, Martin D, Feneley MP, Fatkin D (2004) Defects in nuclear structure and function promote dilated cardiomyopathy in lamin A/C-deficient mice. J Clin Invest 113(3):357–369

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  29. Jahn D, Schramm S, Schnolzer M, Heilmann CJ, de Koster CG, Schutz W, Benavente R, Alsheimer M (2012) A truncated lamin A in the Lmna−/− mouse line: implications for the understanding of laminopathies. Nucleus 3(5):463–474

    Article  PubMed Central  PubMed  Google Scholar 

  30. Kubben N, Voncken JW, Konings G, van Weeghel M, van den Hoogenhof MM, Gijbels M, van Erk A, Schoonderwoerd K, van den Bosch B, Dahlmans V, Calis C, Houten SM, Misteli T, Pinto YM (2011) Post-natal myogenic and adipogenic developmental: defects and metabolic impairment upon loss of A-type lamins. Nucleus 2(3):195–207

    Article  PubMed Central  PubMed  Google Scholar 

  31. van Engelen BG, Muchir A, Hutchison CJ, van der Kooi AJ, Bonne G, Lammens M (2005) The lethal phenotype of a homozygous nonsense mutation in the lamin A/C gene. Neurology 64(2):374–376

    Article  PubMed  Google Scholar 

  32. Boban M, Braun J, Foisner R (2010) Lamins: ‘structure goes cycling’. Biochem Soc Trans 38(Pt 1):301–306

    Article  PubMed  CAS  Google Scholar 

  33. Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP (1986) A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 323(6089):643–646

    Article  PubMed  CAS  Google Scholar 

  34. Dick FA, Rubin SM (2013) Molecular mechanisms underlying RB protein function. Nat Rev Mol Cell Biol 14(5):297–306

    Article  PubMed  CAS  Google Scholar 

  35. Morris EJ, Dyson N (2001) Retinoblastoma protein partners. Adv Cancer Res 82:1–54

    Article  PubMed  CAS  Google Scholar 

  36. Markiewicz E, Dechat T, Foisner R, Quinlan RA, Hutchison CJ (2002) Lamin A/C binding protein LAP2a is required for nuclear anchorage of retinoblastoma protein. Mol Biol Cell 13:4401–4413

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  37. Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotzmann J, Hutchison CJ, Foisner R (2006) Lamina-associated polypeptide 2alpha regulates cell cycle progression and differentiation via the retinoblastoma-E2F pathway. J Cell Biol 173(1):83–93

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  38. Nitta RT, Jameson SA, Kudlow BA, Conlan LA, Kennedy BK (2006) Stabilization of the retinoblastoma protein by A-type nuclear lamins is required for INK4A-mediated cell cycle arrest. Mol Cell Biol 26:5360–5372

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  39. 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 U S A 101:9677–9682

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  40. Naeger LK, Goodwin EC, Hwang ES, DeFilippis RA, Zhang H, DiMaio D (1999) Bovine papillomavirus E2 protein activates a complex growth-inhibitory program in p53 negative HT-3 cervical carcinoma cells that includes repression of cyclin A and cdc25A phosphatase genes and accumulation of hypophosphorylated retinoblastoma protein. Cell Growth Differ 10(6):413–422

    PubMed  CAS  Google Scholar 

  41. Pekovic V, Harborth J, Broers JL, Ramaekers FC, van Engelen B, Lammens M, von Zglinicki T, Foisner R, Hutchison C, Markiewicz E (2007) Nucleoplasmic LAP2{alpha}-lamin A complexes are required to maintain a proliferative state in human fibroblasts. J Cell Biol 176(2):163–172

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  42. Van Berlo JH, Voncken JW, Kubben N, Broers JL, Duisters R, van Leeuwen RE, Crijns HJ, Ramaekers FC, Hutchison CJ, Pinto YM (2005) A-type lamins are essential for TGF-beta1 induced PP2A to dephosphorylate transcription factors. Hum Mol Genet 14(19):2839–2849

    Article  PubMed  Google Scholar 

  43. 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 2(9):e963

    Article  PubMed Central  PubMed  Google Scholar 

  44. Muchir A, Massart C, van Engelen BG, Lammens M, Bonne G, Worman HJ (2006) Proteasome-mediated degradation of integral inner nuclear membrane protein emerin in fibroblasts lacking A-type lamins. Biochem Biophys Res Commun 351(4):1011–1017

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  45. Muralikrishna B, Chaturvedi P, Sinha K, Parnaik VK (2012) Lamin misexpression upregulates three distinct ubiquitin ligase systems that degrade ATR kinase in HeLa cells. Mol Cell Biochem 365(1–2):323–332

    Article  PubMed  CAS  Google Scholar 

  46. Verde P, Casalino L, Talotta F, Yaniv M, Weitzman JB (2007) Deciphering AP-1 function in tumorigenesis: fra-ternizing on target promoters. Cell Cycle 6(21):2633–2639

    Article  PubMed  CAS  Google Scholar 

  47. Ivorra C, Kubicek M, Gonzalez JM, Sanz-Gonzalez SM, Alvarez-Barrientos A, O’Connor JE, Burke B, Andres V (2006) A mechanism of AP-1 suppression through interaction of c-Fos with lamin A/C. Genes Dev 20(3):307–320

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  48. 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

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  49. Muchir A, Wu W, Worman HJ (2010) Mitogen-activated protein kinase inhibitor regulation of heart function and fibrosis in cardiomyopathy caused by lamin A/C gene mutation. Trends Cardiovasc Med 20(7):217–221

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  50. Rodriguez J, Calvo F, Gonzalez JM, Casar B, Andres V, Crespo P (2010) ERK1/2 MAP kinases promote cell cycle entry by rapid, kinase-independent disruption of retinoblastoma-lamin A complexes. J Cell Biol 191(5):967–979

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  51. Ellis DJ, Jenkins H, Whitfield WG, Hutchison CJ (1997) GST-lamin fusion proteins act as dominant negative mutants in Xenopus egg extract and reveal the function of the lamina in DNA replication. J Cell Sci 110:2507–2518

    PubMed  CAS  Google Scholar 

  52. Spann TP, Moir RD, Goldman AE, Stick R, Goldman RD (1997) Disruption of nuclear lamin organization alters the distribution of replication factors and inhibits DNA synthesis. J Cell Biol 136:1201–1212

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  53. Moir RD, Spann TP, Herrmann H, Goldman RD (2000) Disruption of nuclear lamin organization blocks the elongation phase of DNA replication. J Cell Biol 149:1179–1192

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  54. Moir RD, Spann TP, Goldman RD (1994) Dynamic properties of nuclear lamins: lamin B is associated with sites of DNA replication. J Cell Biol 125:1201–1212

    Article  PubMed  CAS  Google Scholar 

  55. Kennedy BK, Barbie DA, Classon M, Dyson N, Harlow E (2000) Nuclear organization of DNA replication in primary mammalian cells. Genes Dev 14:2855–2868

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  56. Singh M, Hunt CR, Pandita RK, Kumar R, Yang CR, Horikoshi N, Bachoo R, Serag S, Story MD, Shay JW, Powell SN, Gupta A, Jeffery J, Pandita S, Chen BP, Deckbar D, Lobrich M, Yang Q, Khanna KK, Worman HJ, Pandita TK (2013) Lamin A/C depletion enhances DNA damage-induced stalled replication fork arrest. Mol Cell Biol 33(6):1210–1222

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  57. Odgren PR, Pratt CH, Mackay CA, Mason-Savas A, Curtain M, Shopland L, Ichicki T, Sundberg JP, Donahue LR (2010) Disheveled hair and ear (Dhe), a spontaneous mouse Lmna mutation modeling human laminopathies. PLoS One 5(4):e9959

    Article  PubMed Central  PubMed  Google Scholar 

  58. Pratt CH, Curtain M, Donahue LR, Shopland LS (2011) Mitotic defects lead to pervasive aneuploidy and accompany loss of RB1 activity in mouse LmnaDhe dermal fibroblasts. PLoS One 6(3):e18065

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  59. Campisi J (2013) Aging, cellular senescence, and cancer. Annu Rev Physiol 75:685–705

    Article  PubMed  CAS  Google Scholar 

  60. 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 U S A 104(12):4955–4960

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  61. Cao K, Capell BC, Erdos MR, Djabali K, Collins FS (2007) A lamin A protein isoform overexpressed in Hutchinson-Gilford progeria syndrome interferes with mitosis in progeria and normal cells. Proc Natl Acad Sci U S A 104(12):4949–4954

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  62. Liu B, Wang J, Chan KM, Tjia WM, Deng W, Guan X, Huang JD, Li KM, Chau PY, Chen DJ, Pei D, Pendas AM, Cadinanos J, Lopez-Otin C, Tse HF, Hutchison C, Chen J, Cao Y, Cheah KS, Tryggvason K, Zhou Z (2005) Genomic instability in laminopathy-based premature aging. Nat Med 11(7):780–785

    Article  PubMed  CAS  Google Scholar 

  63. Varela I, Cadinanos J, Pendas AM, Gutierrez-Fernandez A, Folgueras AR, Sanchez LM, Zhou Z, Rodriguez FJ, Stewart CL, Vega JA, Tryggvason K, Freije JM, Lopez-Otin C (2005) Accelerated ageing in mice deficient in Zmpste24 protease is linked to p53 signalling activation. Nature 437(7058):564–568

    Article  PubMed  CAS  Google Scholar 

  64. Kudlow BA, Stanfel MN, Burtner CR, Johnston ED, Kennedy BK (2008) Suppression of proliferative defects associated with processing-defective lamin A mutants by hTERT or inactivation of p53. Mol Biol Cell 19(12):5238–5248

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  65. Marji J, O’Donoghue SI, McClintock D, Satagopam VP, Schneider R, Ratner D, Worman HJ, Gordon LB, Djabali K (2010) Defective lamin A-Rb signaling in Hutchinson-Gilford progeria syndrome and reversal by farnesyltransferase inhibition. PLoS One 5(6):e11132

    Article  PubMed Central  PubMed  Google Scholar 

  66. King CR, Lemmer J, Campbell JR, Atkins AR (1978) Osteosarcoma in a patient with Hutchinson-Gilford progeria. J Med Genet 15(6):481–484

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  67. Shalev SA, De Sandre-Giovannoli A, Shani AA, Levy N (2007) An association of Hutchinson-Gilford progeria and malignancy. Am J Med Genet A 143A(16):1821–1826

    Article  PubMed  CAS  Google Scholar 

  68. Wadayama B, Toguchida J, Shimizu T, Ishizaki K, Sasaki MS, Kotoura Y, Yamamuro T (1994) Mutation spectrum of the retinoblastoma gene in osteosarcomas. Cancer Res 54(11):3042–3048

    PubMed  CAS  Google Scholar 

  69. 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 161(7):1599–1611

    Article  CAS  Google Scholar 

  70. Moiseeva O, Bourdeau V, Vernier M, Dabauvalle MC, Ferbeyre G (2011) Retinoblastoma-independent regulation of cell proliferation and senescence by the p53-p21 axis in lamin A/C-depleted cells. Aging Cell 10(5):789–797

    Article  PubMed  CAS  Google Scholar 

  71. Jung YS, Lee SJ, Lee SH, Chung JY, Jung YJ, Hwang SH, Ha NC, Park BJ (2013) Loss of VHL promotes progerin expression, leading to impaired p14/ARF function and suppression of p53 activity. Cell Cycle 12(14)

    Google Scholar 

  72. Gonzalez-Suarez I, Gonzalo S (2010) Nurturing the genome: A-type lamins preserve genomic stability. Nucleus 1(2):129–135

    Article  PubMed Central  PubMed  Google Scholar 

  73. Allsopp RC, Vaziri H, Patterson C, Goldstein S, Younglai EV, Futcher AB, Greider CW, Harley CB (1992) Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci U S A 89(21):10114–10118

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  74. Shoeman RL, Wadle S, Scherbarth A, Traub P (1988) The binding in vitro of the intermediate filament protein vimentin to synthetic oligonucleotides containing telomere sequences. J Biol Chem 263(35):18744–18749

    PubMed  CAS  Google Scholar 

  75. de Lange T (1992) Human telomeres are attached to the nuclear matrix. EMBO J 11(2):717–724

    PubMed Central  PubMed  Google Scholar 

  76. Luderus ME, van Steensel B, Chong L, Sibon OC, Cremers FF, de Lange T (1996) Structure, subnuclear distribution, and nuclear matrix association of the mammalian telomeric complex. J Cell Biol 135(4):867–881

    Article  PubMed  CAS  Google Scholar 

  77. Raz V, Vermolen BJ, Garini Y, Onderwater JJ, Mommaas-Kienhuis MA, Koster AJ, Young IT, Tanke H, Dirks RW (2008) The nuclear lamina promotes telomere aggregation and centromere peripheral localization during senescence of human mesenchymal stem cells. J Cell Sci 121(Pt 24):4018–4028

    Article  PubMed  CAS  Google Scholar 

  78. Gonzalez-Suarez I, Redwood AB, Perkins SM, Vermolen B, Lichtensztejin D, Grotsky DA, Morgado-Palacin L, Gapud EJ, Sleckman BP, Sullivan T, Sage J, Stewart CL, Mai S, Gonzalo S (2009) Novel roles for A-type lamins in telomere biology and the DNA damage response pathway. EMBO J 28(16):2414–2427

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  79. Gonzalo S, Garcia-Cao M, Fraga MF, Schotta G, Peters AH, Cotter SE, Eguia R, Dean DC, Esteller M, Jenuwein T, Blasco MA (2005) Role of the RB1 family in stabilizing histone methylation at constitutive heterochromatin. Nat Cell Biol 7(4):420–428

    Article  PubMed  CAS  Google Scholar 

  80. Gonzalo S, Blasco MA (2005) Role of Rb family in the epigenetic definition of chromatin. Cell Cycle 4(6):752–755

    Article  PubMed  CAS  Google Scholar 

  81. Benetti R, Gonzalo S, Jaco I, Schotta G, Klatt P, Jenuwein T, Blasco MA (2007) Suv4-20h deficiency results in telomere elongation and derepression of telomere recombination. J Cell Biol 178(6):925–936

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  82. Garcia-Cao M, O’Sullivan R, Peters AH, Jenuwein T, Blasco MA (2004) Epigenetic regulation of telomere length in mammalian cells by the Suv39h1 and Suv39h2 histone methyltransferases. Nat Genet 36(1):94–99

    Article  PubMed  CAS  Google Scholar 

  83. Gonzalez-Suarez I, Redwood AB, Gonzalo S (2009) Loss of A-type lamins and genomic instability. Cell Cycle 8(23):3860–3865

    Article  PubMed  CAS  Google Scholar 

  84. Shumaker DK, Dechat T, Kohlmaier A, Adam SA, Bozovsky MR, Erdos MR, Eriksson M, Goldman AE, Khuon S, Collins FS, Jenuwein T, Goldman RD (2006) Mutant nuclear lamin A leads to progressive alterations of epigenetic control in premature aging. Proc Natl Acad Sci U S A 103(23):8703–8708

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  85. Benson EK, Lee SW, Aaronson SA (2010) Role of progerin-induced telomere dysfunction in HGPS premature cellular senescence. J Cell Sci 123(Pt 15):2605–2612

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  86. 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

    Article  PubMed  CAS  Google Scholar 

  87. Barascu A, Le Chalony C, Pennarun G, Genet D, Zaarour N, Bertrand P (2012) Oxidative stress alters nuclear shape through lamins dysregulation: a route to senescence. Nucleus 3(5):411–417

    Article  PubMed Central  PubMed  Google Scholar 

  88. Freund A, Laberge RM, Demaria M, Campisi J (2012) Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell 23(11):2066–2075

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  89. Dreesen O, Chojnowski A, Ong PF, Zhao TY, Common JE, Lunny D, Lane EB, Lee SJ, Vardy LA, Stewart CL, Colman A (2013) Lamin B1 fluctuations have differential effects on cellular proliferation and senescence. J Cell Biol 200(5):605–617

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  90. Padiath QS, Saigoh K, Schiffmann R, Asahara H, Yamada T, Koeppen A, Hogan K, Ptacek LJ, Fu YH (2006) Lamin B1 duplications cause autosomal dominant leukodystrophy. Nat Genet 38(10):1114–1123

    Article  PubMed  CAS  Google Scholar 

  91. Barascu A, Le Chalony C, Pennarun G, Genet D, Imam N, Lopez B, Bertrand P (2012) Oxidative stress induces an ATM-independent senescence pathway through p38 MAPK-mediated lamin B1 accumulation. EMBO J 31(5):1080–1094

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  92. Candelario J, Sudhakar S, Navarro S, Reddy S, Comai L (2008) Perturbation of wild-type lamin A metabolism results in a progeroid phenotype. Aging Cell 7(3):355–367

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  93. Harman D (1956) Aging: a theory based on free radical and radiation chemistry. J Gerontol 11:298–300

    Article  PubMed  CAS  Google Scholar 

  94. Pekovic V, Gibbs-Seymour I, Markiewicz E, Alzoghaibi F, Benham AM, Edwards R, Wenhert M, von Zglinicki T, Hutchison CJ (2011) Conserved cysteine residues in the mammalian lamin A tail are essential for cellular responses to ROS generation. Aging Cell 10(6):1067–1079

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Lamin-related research in the lab of B.K.K. is supported by a grant from the National Institute of Aging (R01 AG024287).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Brian K. Kennedy Ph.D. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this chapter

Cite this chapter

Kennedy, B.K., Pennypacker, J.K. (2014). RB and Lamins in Cell Cycle Regulation and Aging. In: Schirmer, E., de las Heras, J. (eds) Cancer Biology and the Nuclear Envelope. Advances in Experimental Medicine and Biology, vol 773. Springer, New York, NY. https://doi.org/10.1007/978-1-4899-8032-8_6

Download citation

Publish with us

Policies and ethics