Display options
Share it on

Cell Mol Life Sci. 2022 Jan 04;79(1):23. doi: 10.1007/s00018-021-04099-9.

Transient nuclear lamin A/C accretion aids in recovery from vapor nanobubble-induced permeabilisation of the plasma membrane.

Cellular and molecular life sciences : CMLS

Gaëlle Houthaeve, Gerardo García-Díaz Barriga, Stephan Stremersch, Herlinde De Keersmaecker, Juan Fraire, Jo Vandesompele, Pieter Mestdagh, Stefaan De Smedt, Kevin Braeckmans, Winnok H De Vos

Affiliations

  1. Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium.
  2. Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000, Ghent, Belgium.
  3. Centre for Advanced Light Microscopy, Ghent University, 9000, Ghent, Belgium.
  4. Center for Medical Genetics, Ghent University, 9000, Ghent, Belgium.
  5. Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium. [email protected].

PMID: 34984553 DOI: 10.1007/s00018-021-04099-9

Abstract

Vapor nanobubble (VNB) photoporation is a physical method for intracellular delivery that has gained significant interest in the past decade. It has successfully been used to introduce molecular cargo of diverse nature into different cell types with high throughput and minimal cytotoxicity. For translational purposes, it is important to understand whether and how photoporation affects cell homeostasis. To obtain a comprehensive view on the transcriptional rewiring that takes place after VNB photoporation, we performed a longitudinal shotgun RNA-sequencing experiment. Six hours after photoporation, we found a marked upregulation of LMNA transcripts as well as their protein products, the A-type lamins. At the same time point, we observed a significant increase in several heterochromatin marks, suggesting a global stiffening of the nucleus. These molecular features vanished 24 h after photoporation. Since VNB-induced chromatin condensation was prolonged in LMNA knockout cells, A-type lamins may be required for restoring the nucleus to its original state. Selective depletion of A-type lamins reduced cell viability after VNB photoporation, while pharmacological stimulation of LMNA transcription increased the percentage of successfully transfected cells that survived after photoporation. Therefore, our results suggest that cells respond to VNB photoporation by temporary upregulation of A-type lamins to facilitate their recovery.

© 2022. The Author(s).

Keywords: A-type lamins; Chromatin; Gold nanoparticles; Photoporation; Plasma membrane; Vapor nanobubbles

References

  1. Morshedi Rad D et al (2021) A comprehensive review on intracellular delivery. Adv Mater 33(13):e2005363 - PubMed
  2. Stewart MP, Langer R, Jensen KF (2018) Intracellular delivery by membrane disruption: mechanisms, strategies, and concepts. Chem Rev 118(16):7409–7531 - PubMed
  3. Howarth JL, Lee YB, Uney JB (2010) Using viral vectors as gene transfer tools (Cell Biology and Toxicology Special ssue: ETCS-UK 1 day meeting on genetic manipulation of cells). Cell Biol Toxicol 26(1):1–20 - PubMed
  4. Thomas CE, Ehrhardt A, Kay MA (2003) Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet 4(5):346–358 - PubMed
  5. Meacham JM et al (2014) Physical methods for intracellular delivery: practical aspects from laboratory use to industrial-scale processing. J Lab Autom 19(1):1–18 - PubMed
  6. Hapala I (1997) Breaking the barrier: methods for reversible permeabilization of cellular membranes. Crit Rev Biotechnol 17(2):105–122 - PubMed
  7. Mellott AJ, Forrest ML, Detamore MS (2013) Physical non-viral gene delivery methods for tissue engineering. Ann Biomed Eng 41(3):446–468 - PubMed
  8. Xiong R et al (2016) Laser-assisted photoporation: fundamentals, technological advances and applications. Adv Phys X 1(4):596–620 - PubMed
  9. Urban AS et al (2009) Controlled nanometric phase transitions of phospholipid membranes by plasmonic heating of single gold nanoparticles. Nano Lett 9(8):2903–2908 - PubMed
  10. Delcea M et al (2012) Nanoplasmonics for dual-molecule release through nanopores in the membrane of red blood cells. ACS Nano 6(5):4169–4180 - PubMed
  11. Xiong R et al (2014) Comparison of gold nanoparticle mediated photoporation: vapor nanobubbles outperform direct heating for delivering macromolecules in live cells. ACS Nano 8(6):6288–6296 - PubMed
  12. Xiong R et al (2016) Cytosolic delivery of nanolabels prevents their asymmetric inheritance and enables extended quantitative in vivo cell imaging. Nano Lett. https://doi.org/10.1021/acs.nanolett.6b01411 - PubMed
  13. Xiong R et al (2017) Fast spatial-selective delivery into live cells. J Control Release 266(2):198–204 - PubMed
  14. Wayteck L et al (2017) Comparing photoporation and nucleofection for delivery of small interfering RNA to cytotoxic T cells. J Control Release 267:154–162 - PubMed
  15. Raes L et al (2020) Intracellular delivery of mRNA in adherent and suspension cells by vapor nanobubble photoporation. Nanomicro Lett 12(1):185 - PubMed
  16. Schomaker M et al (2015) Characterization of nanoparticle mediated laser transfection by femtosecond laser pulses for applications in molecular medicine. J Nanobiotechnology 13:10 - PubMed
  17. Lukianova-Hleb EY et al (2012) Cell-specific transmembrane injection of molecular cargo with gold nanoparticle-generated transient plasmonic nanobubbles. Biomaterials 33(21):5441–5450 - PubMed
  18. Santra TS et al (2020) Near-infrared nanosecond-pulsed laser-activated highly efficient intracellular delivery mediated by nano-corrugated mushroom-shaped gold-coated polystyrene nanoparticles. Nanoscale 12(22):12057–12067 - PubMed
  19. Van Hoecke L et al (2019) Delivery of mixed-lineage kinase domain-like protein by vapor nanobubble photoporation induces necroptotic-like cell death in tumor cells. Int J Mol Sci 20(17):4254 - PubMed
  20. Yao C et al (2020) Cancer cell-specific protein delivery by optoporation with laser-irradiated gold nanorods. J Biophotonics 13(7):e202000017 - PubMed
  21. Schomaker M et al (2015) Biophysical effects in off-resonant gold nanoparticle mediated (GNOME) laser transfection of cell lines, primary- and stem cells using fs laser pulses. J Biophotonics 8(8):646–658 - PubMed
  22. Chen X et al (2014) Single-site sonoporation disrupts actin cytoskeleton organization. J R Soc Interface 11(95):20140071 - PubMed
  23. Wang M et al (2018) Sonoporation-induced cell membrane permeabilization and cytoskeleton disassembly at varied acoustic and microbubble-cell parameters. Sci Rep 8(1):3885 - PubMed
  24. Fan P et al (2017) Cell-cycle-specific cellular responses to sonoporation. Theranostics 7(19):4894–4908 - PubMed
  25. Juffermans LJ et al (2009) Ultrasound and microbubble-induced intra- and intercellular bioeffects in primary endothelial cells. Ultrasound Med Biol 35(11):1917–1927 - PubMed
  26. Kumon RE et al (2009) Spatiotemporal effects of sonoporation measured by real-time calcium imaging. Ultrasound Med Biol 35(3):494–506 - PubMed
  27. Zhong W et al (2013) Induction of endoplasmic reticulum stress by sonoporation: linkage to mitochondria-mediated apoptosis initiation. Ultrasound Med Biol 39(12):2382–2392 - PubMed
  28. Chen X, Wan JM, Yu AC (2013) Sonoporation as a cellular stress: induction of morphological repression and developmental delays. Ultrasound Med Biol 39(6):1075–1086 - PubMed
  29. Zhong W et al (2011) Sonoporation induces apoptosis and cell cycle arrest in human promyelocytic leukemia cells. Ultrasound Med Biol 37(12):2149–2159 - PubMed
  30. Schmiderer L et al (2020) Efficient and nontoxic biomolecule delivery to primary human hematopoietic stem cells using nanostraws. Proc Natl Acad Sci USA 117(35):21267–21273 - PubMed
  31. DiTommaso T et al (2018) Cell engineering with microfluidic squeezing preserves functionality of primary immune cells in vivo. Proc Natl Acad Sci USA 115(46):E10907–E10914 - PubMed
  32. Kalies S et al (2015) Investigation of biophysical mechanisms in gold nanoparticle mediated laser manipulation of cells using a multimodal holographic and fluorescence imaging setup. PLoS One 10(4):e0124052 - PubMed
  33. Saklayen N et al (2017) Analysis of poration-induced changes in cells from laser-activated plasmonic substrates. Biomed Opt Express 8(10):4756–4771 - PubMed
  34. Johannsmeier S et al (2018) Gold nanoparticle-mediated laser stimulation induces a complex stress response in neuronal cells. Sci Rep 8(1):6533 - PubMed
  35. Fraire JC et al (2020) Vapor nanobubble is the more reliable photothermal mechanism for inducing endosomal escape of siRNA without disturbing cell homeostasis. J Control Release 319:262–275 - PubMed
  36. Lin F, Worman HJ (1993) Structural organization of the human gene encoding nuclear lamin A and nuclear lamin C. J Biol Chem 268(22):16321–16326 - PubMed
  37. Mascetti G et al (1996) Effect of fixatives on calf thymocytes chromatin as analyzed by 3D high-resolution fluorescence microscopy. Cytometry 23(2):110–119 - PubMed
  38. Vergani L, Mascetti G, Nicolini C (2001) Changes of nuclear structure induced by increasing temperatures. J Biomol Struct Dyn 18(4):535–544 - PubMed
  39. Vergani L, Grattarola M, Nicolini C (2004) Modifications of chromatin structure and gene expression following induced alterations of cellular shape. Int J Biochem Cell Biol 36(8):1447–1461 - PubMed
  40. Isaacson SA, McQueen DM, Peskin CS (2011) The influence of volume exclusion by chromatin on the time required to find specific DNA binding sites by diffusion. Proc Natl Acad Sci USA 108(9):3815–3820 - PubMed
  41. Versaevel M, Grevesse T, Gabriele S (2012) Spatial coordination between cell and nuclear shape within micropatterned endothelial cells. Nat Commun 3:671 - PubMed
  42. Irianto J et al (2013) Osmotic challenge drives rapid and reversible chromatin condensation in chondrocytes. Biophys J 104(4):759–769 - PubMed
  43. Wiles ET, Selker EU (2017) H3K27 methylation: a promiscuous repressive chromatin mark. Curr Opin Genet Dev 43:31–37 - PubMed
  44. Huang C, Zhu B (2018) Roles of H3K36-specific histone methyltransferases in transcription: antagonizing silencing and safeguarding transcription fidelity. Biophys Rep 4(4):170–177 - PubMed
  45. van Steensel B, Belmont AS (2017) Lamina-associated domains: links with chromosome architecture, heterochromatin, and gene repression. Cell 169(5):780–791 - PubMed
  46. Briand N, Collas P (2020) Lamina-associated domains: peripheral matters and internal affairs. Genome Biol 21(1):85 - PubMed
  47. Robijns J et al (2016) In silico synchronization reveals regulators of nuclear ruptures in lamin A/C deficient model cells. Sci Rep 6:30325 - PubMed
  48. Swift J et al (2013) Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341(6149):1240104 - PubMed
  49. Stephens AD et al (2018) Chromatin histone modifications and rigidity affect nuclear morphology independent of lamins. Mol Biol Cell 29(2):220–233 - PubMed
  50. Tariq Z et al (2017) Lamin A and microtubules collaborate to maintain nuclear morphology. Nucleus 8(4):433–446 - PubMed
  51. Gerlitz G, Reiner O, Bustin M (2013) Microtubule dynamics alter the interphase nucleus. Cell Mol Life Sci 70(7):1255–1268 - PubMed
  52. Eden E et al (2011) Proteome half-life dynamics in living human cells. Science 331(6018):764–768 - PubMed
  53. Lammerding J et al (2004) Lamin A/C deficiency causes defective nuclear mechanics and mechanotransduction. J Clin Invest 113(3):370–378 - PubMed
  54. Cho S, Irianto J, Discher DE (2017) Mechanosensing by the nucleus: From pathways to scaling relationships. J Cell Biol 216(2):305–315 - PubMed
  55. Stephens AD et al (2017) Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus. Mol Biol Cell 28(14):1984–1996 - PubMed
  56. Hobson CM et al (2020) Correlating nuclear morphology and external force with combined atomic force microscopy and light sheet imaging separates roles of chromatin and lamin A/C in nuclear mechanics. Mol Biol Cell 31(16):1788–1801 - PubMed
  57. Lukianova-Hleb E et al (2010) Plasmonic nanobubbles as transient vapor nanobubbles generated around plasmonic nanoparticles. ACS Nano 4(4):2109–2123 - PubMed
  58. Booth EA et al (2015) Nuclear stiffening and chromatin softening with progerin expression leads to an attenuated nuclear response to force. Soft Matter 11(32):6412–6418 - PubMed
  59. Damodaran K et al (2018) Compressive force induces reversible chromatin condensation and cell geometry-dependent transcriptional response. Mol Biol Cell 29(25):3039–3051 - PubMed
  60. Stephens AD (2020) Chromatin rigidity provides mechanical and genome protection. Mutat Res 821:111712 - PubMed
  61. Nava MM et al (2020) Heterochromatin-driven nuclear softening protects the genome against mechanical stress-induced damage. Cell 181(4):800-817.e22 - PubMed
  62. Miroshnikova YA, Nava MM, Wickstrom SA (2017) Emerging roles of mechanical forces in chromatin regulation. J Cell Sci 130(14):2243–2250 - PubMed
  63. Dechat T, Gesson K, Foisner R (2010) Lamina-independent lamins in the nuclear interior serve important functions. Cold Spring Harb Symp Quant Biol 75:533–543 - PubMed
  64. Gesson K, Vidak S, Foisner R (2014) Lamina-associated polypeptide (LAP)2alpha and nucleoplasmic lamins in adult stem cell regulation and disease. Semin Cell Dev Biol 29:116–124 - PubMed
  65. Bronshtein I et al (2015) Loss of lamin A function increases chromatin dynamics in the nuclear interior. Nat Commun 6:8044 - PubMed
  66. Belin BJ et al (2013) Visualization of actin filaments and monomers in somatic cell nuclei. Mol Biol Cell 24(7):982–994 - PubMed
  67. Le HQ et al (2016) Mechanical regulation of transcription controls Polycomb-mediated gene silencing during lineage commitment. Nat Cell Biol 18(8):864–875 - PubMed
  68. Vermeulen LMP et al (2018) Photothermally triggered endosomal escape and its influence on transfection efficiency of gold-functionalized JetPEI/pDNA nanoparticles. Int J Mol Sci 19(8):2400 - PubMed
  69. Lapotko D (2009) Optical excitation and detection of vapor bubbles around plasmonic nanoparticles. Opt Express 17(4):2538–2556 - PubMed
  70. Zhou Y et al (2019) Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun 10(1):1523 - PubMed
  71. De Vos WH et al (2010) High content image cytometry in the context of subnuclear organization. Cytometry A 77(1):64–75 - PubMed
  72. De Puysseleyr L, De Puysseleyr K, Vanrompay D, De Vos W (2017) Quantyifying the growth of Chlamydia suis in cell culture using high-content microscopy. Microsc Res Tech 80(4):350–356. https://doi.org/10.1002/jemt.22799 - PubMed

Publication Types

Grant support