Summary

Nuclei Isolation and Super-Resolution Structured Illumination Microscopy for Examining Nucleoporin Alterations in Human Neurodegeneration

Published: September 10, 2021
doi:

Summary

This protocol describes an optimized workflow for nuclei isolation and super-resolution structured illumination microscopy to evaluate individual nucleoporins within the nucleoplasm and NPCs in induced pluripotent stem cell derived neurons and postmortem human tissues.

Abstract

The nuclear pore complex (NPC) is a complex macromolecular structure comprised of multiple copies of ~30 different nucleoporin proteins (Nups). Collectively, these Nups function to regulate genome organization, gene expression, and nucleocytoplasmic transport (NCT). Recently, defects in NCT and alterations to specific Nups have been identified as early and prominent pathologies in multiple neurodegenerative diseases, including Amyotrophic Lateral Sclerosis (ALS), Alzheimer's Disease (AD)/Frontotemporal Dementia (FTD), and Huntington's Disease (HD). Advances in both light and electron microscopy allow for a thorough examination of sub-cellular structures, including the NPC and its Nup constituents, with increased precision and resolution. Of the commonly used techniques, super-resolution structured illumination microscopy (SIM) affords the unparalleled opportunity to study the localization and expression of individual Nups using conventional antibody-based labeling strategies. Isolation of nuclei prior to SIM enables the visualization of individual Nup proteins within the NPC and nucleoplasm in fully and accurately reconstructed 3D space. This protocol describes a procedure for nuclei isolation and SIM to evaluate Nup expression and distribution in human iPSC-derived CNS cells and postmortem tissues.

Introduction

The prevalence of age-related neurodegenerative diseases is increasing as the population ages1. While the genetic underpinnings and pathologic hallmarks are well characterized, the precise molecular events leading to neuronal injury remain poorly understood2,3,4,5,6,7,8,9,10,11,12. Recently, a G4C2 hexanucleotide repeat expansion in the first intron of the C9orf72 gene was identified as the most common genetic cause of the related neurodegenerative diseases Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD)13,14. Several studies now support a central role for disruptions in the nuclear transport machinery, including nuclear pore complexes (NPCs) and nuclear transport receptors (NTRs, karyopherins), as being causative of C9orf72 ALS15,16. In non-dividing cells within the rat brain, scaffold nucleoporins (Nups) are extremely long-lived. As a result, alterations in NPCs and NCT have been reported during aging17,18,19,20. Moreover, some nucleoporins or transportins, when mutated, are linked to specific neurological diseases21,22. For example, mutations in Nup62 have been linked to Infantile Bilateral Striatal Necrosis (IBSN), a neurological disorder affecting the caudate nucleus and putamen23; mutations in Gle1 have been implicated in the fetal motor neuron disease Human Lethal Congenital Contracture Syndrome-1 (LCCS1)24; and mutations in Aladin are causative of Triple-A Syndrome25. Alterations in functional NCT are exacerbated in age-related neurodegenerative diseases such as ALS, Huntington's Disease (HD), and Alzheimer's Disease (AD)16,26,27,28,29,30,31. In addition, specific Nups and NTRs have been reported as modifiers of C9orf72 mediated toxicity in the Drosophila eye28 or biochemically modify the aggregation state of disease-linked proteins such as FUS and tau27,32,33,34. Collectively, these early studies suggest that altered NCT may be a primary and early pathological feature of ALS and FTD. Studies in overexpression-based model system have suggested that mislocalization of specific Nups and karyopherins may impact NCT16,35,36,37,38. However, these pathology studies do not actually link cytoplasmic accumulations of NPC proteins to defects in the structure or function of the NPC. For example, this pathology may simply reflect the dysregulation of cytoplasmic pools of Nup proteins with little impact on NPC composition and function. In contrast, a recent study employing super resolution structured illumination microscopy (SIM) demonstrates the emergence of a significant injury to the NPC itself characterized by reduction in specific Nup levels within the nucleoplasm and NPCs of human C9orf72 ALS/FTD neurons ultimately leading to altered NPC function as an early initiating event in pathogenic disease cascades15.

The passage of macromolecules between the nucleus and cytoplasm is governed by the nuclear pore complex (NPC). The NPC is a large macromolecular complex embedded in the nuclear envelope comprised of multiple copies of 30 nucleoporin proteins (Nups)39,40,41. Although Nup stoichiometry varies among cell types42,43,44, maintenance of overall NPC composition is critical for NCT, genome organization, and overall cellular viability39,41,45,46. As a result, altered NPC composition and subsequent defects in functional transport are likely to impact a myriad of downstream cellular functions. The Nup constituents of the NPC are highly organized into multiple subcomplexes, including the cytoplasmic ring and filaments, central channel, outer ring, inner ring, transmembrane ring, and nuclear basket. Collectively, scaffold Nups of the inner, outer, and transmembrane rings anchor NPCs within the nuclear envelope and provide anchor points for Nups of the cytoplasmic ring, central channel, and nuclear basket. While small molecules (<40-60 kD) can passively diffuse through the NPC, the active transport of larger cargoes is facilitated by interactions between nuclear transport receptors (NTRs, karyopherins) and the FG Nups of the cytoplasmic filaments, central channel, and nuclear basket39,40,41,45. Also, a handful of Nups can additionally function outside of the NPC, within the nucleoplasm, to regulate gene expression46,47.

Given that the lateral dimension of a single human NPC is approximately 100-120 nm40, standard widefield or confocal microscopy is insufficient to resolve individual NPCs48. Electron microscopy (EM) techniques such as TEM or SEM are often used to evaluate the overall structure of NPCs39,40. Despite the advantages of these techniques for resolving NPC ultrastructure, they are less commonly used to evaluate the presence of individual Nup proteins within the NPC. The technical limitations of combining antibody or tag-based labeling with these state-of-the-art technologies, TEM and SEM, do not always allow for an accurate and reliable assessment of individual Nups themselves within NPCs or the nucleoplasm. Further, these techniques can be technically challenging and are not yet widely accessible to all researchers. However, recent advances in light and fluorescence microscopy have increased the accessibility of super-resolution imaging technologies. Specifically, SIM affords the unparalleled opportunity to image individual Nups with a resolution that approaches the lateral dimensions of one human NPC40,48,49,50,51. In contrast to other super-resolution approaches such as stochastic optical reconstruction microscopy (STORM) and stimulated emission depletion (STED), SIM is compatible with conventional antibody-based immunostaining49. Thus, SIM allows for a comprehensive analysis of all Nups for which a specific Nup antibody is available. The ability to sample and image multiple different Nups in the same preparation provides significant advantages to other imaging methods when surveying the many proteins that comprise the NPC. The following procedure details an optimized protocol for evaluating individual Nup components of the NPC using nuclei isolated from induced pluripotent stem cell (iPSC) derived neurons (iPSNs) and postmortem human central nervous system (CNS) tissues.

Protocol

All blood samples for iPSC generation and autopsied tissue collections are approved by Johns Hopkins IRB with Johns Hopkins ethics oversight. All patient information is HIPPA compliant. The following protocol adheres to all Johns Hopkins biosafety procedures. 1. Preparation of slides for immunostaining and imaging Position a positively charged glass microscope slide in an empty cytofunnel and draw a circle with a hydrophobic barrier pen to outline an area to deposit the nuclei.</li…

Representative Results

To examine the NPC and nucleoplasmic distribution and expression of POM121 in human neuronal nuclei, control, and C9orf72 iPSNs were differentiated as previously described15. Postmortem human motor cortex and day 32 iPSNs were lysed and subjected to nuclei isolation and immunostaining as described above. NeuN positive isolated nuclei were imaged by super-resolution structured illumination microscopy (SIM) using a super-resolution structured illumination microscope (Zeiss) and processed using defau…

Discussion

Given the recent identification of NCT deficits as an early and prominent phenomenon in multiple neurodegenerative diseases16,27,28,30,31, there exists a critical need to thoroughly examine the mechanism by which this pathology occurs. As the NPC and its individual Nup proteins critically control functional NCT39,<sup class="…

Declarações

The authors have nothing to disclose.

Acknowledgements

Postmortem human CNS tissues were provided by the Johns Hopkins ALS Autopsy Bank and the Target ALS Postmortem Tissue Core. This work was supported by the ALSA Milton Safenowitz Postdoctoral Fellowship (ANC), as well as funding from NIH-NINDS, Department of Defense, ALS Association, Muscular Dystrophy Association, F Prime, The Robert Packard Center for ALS Research Answer ALS Program, and the Chan Zuckerberg Initiative.

Materials

50 mL conical tubes Fisher Scientific 14-959-49A
Beckman Ultracentrifuge Beckman Coulter
Cell Scrapers Sarstedt 83.183
Collagen Advanced Biomatrix 5005
Coverslips MatTek PCS-170-1818
Cytofunnel Thermo Fisher Scientific A78710020
Cytospin 4 Fisher Scientific A78300003
Dounce Homogenizers DWK Life Sciences 357542
DTT Sigma Aldrich D0632
Eppendorf tubes Fisher Scientific 05-408-129
Goat Anti-Chicken Alexa 647 Thermo Fisher Scientific A-21449
Goat Anti-Mouse Alexa 488 Thermo Fisher Scientific A-11029
Goat Anti-Mouse Alexa 568 Thermo Fisher Scientific A-11031
Goat Anti-Mouse Alexa 647 Thermo Fisher Scientific A-21236
Goat Anti-Rabbit Alexa 488 Thermo Fisher Scientific A-11034
Goat Anti-Rabbit Alexa 568 Thermo Fisher Scientific A-11036
Goat Anti-Rabbit Alexa 647 Thermo Fisher Scientific A-21245
Goat Anti-Rat Alexa 488 Thermo Fisher Scientific A-11006
Goat Anti-Rat Alexa 568 Thermo Fisher Scientific A-11077
Goat Anti-Rat Alexa 647 Thermo Fisher Scientific A-21247
Hemacytometer Fisher Scientific 267110
Microscope Slides Fisher Scientific 12-550-15
Normal Goat Serum Vector Labs S-1000
Nuclei PURE Prep Nuclei Isolation Kit Sigma Aldrich NUC201 Contains Lysis Buffer, 10% Triton X-100, 2 M Sucrose Gradient, Sucrose Cushion Solution, and Nuclei Storage Buffer; Referenced in protocol as "nuclei isolation kit"
PBS Thermo Fisher Scientific 10010023
PFA Electron Microscopy Sciences 15714-S
Prolong Gold Antifade Invitrogen P36930 Referenced in protocol as "hard mount antifade mounting media"
SW 32 Ti Ultracentrifuge Rotor Beckman Coulter 369694 Referenced in protocol as "ultracentrifuge rotor"
Triton X-100 Sigma Aldrich T9284
Trypan Blue Thermo Fisher Scientific 15-250-061
Ultracentrifuge Tubes Beckman Coulter 344058
Nucleoporin Primary Antibodies Primary antibodies suitable for immunofluorescent detection of invidual nucleoporins are available from multiple companies 

Referências

  1. Hou, Y., et al. Ageing as a risk factor for neurodegenerative disease. Nature Reviews. Neurology. 15 (10), 565-581 (2019).
  2. Kim, G., Gautier, O., Tassoni-Tsuchida, E., Ma, X. R., Gitler, A. D. ALS genetics: Gains, losses, and implications for future therapies. Neuron. 108 (5), 822-842 (2020).
  3. Bang, J., Spina, S., Miller, B. L. Frontotemporal dementia. Lancet. 386 (10004), 1672-1682 (2015).
  4. Blauwendraat, C., Nalls, M. A., Singleton, A. B. The genetic architecture of Parkinson’s disease. The Lancet Neurology. 19 (2), 170-178 (2020).
  5. Cacace, R., Sleegers, K., Van Broeckhoven, C. Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimer’s & Dementia. 12 (6), 733-748 (2016).
  6. Di Resta, C., Ferrari, M. New molecular approaches to Alzheimer’s disease. Clinical Biochemistry. 72, 81-86 (2019).
  7. Karch, C. M., Cruchaga, C., Goate, A. M. Alzheimer’s disease genetics: from the bench to the clinic. Neuron. 83 (1), 11-26 (2014).
  8. Kovacs, G. G. Molecular pathology of neurodegenerative diseases: principles and practice. Journal of Clinical Pathology. 72 (11), 725-735 (2019).
  9. McColgan, P., Tabrizi, S. J. Huntington’s disease: a clinical review. European Journal of Neurology. 25 (1), 24-34 (2018).
  10. Ross, C. A., Tabrizi, S. J. Huntington’s disease: from molecular pathogenesis to clinical treatment. The Lancet. Neurology. 10 (1), 83-98 (2011).
  11. Dugger, B. N., Dickson, D. W. Pathology of neurodegenerative diseases. Cold Spring Harbor Perspectives in Biology. 9 (7), 028035 (2017).
  12. Ross, C. A., Poirier, M. A. Protein aggregation and neurodegenerative disease. Nature Medicine. 10, 10-17 (2004).
  13. DeJesus-Hernandez, M., et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 72 (2), 245-256 (2011).
  14. Renton, A. E., et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 72 (2), 257-268 (2011).
  15. Coyne, A. N., et al. G(4)C(2) repeat RNA initiates a POM121-mediated reduction in specific nucleoporins in C9orf72 ALS/FTD. Neuron. 107 (4), 1124-1140 (2020).
  16. Zhang, K., et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature. 525 (7567), 56-61 (2015).
  17. D’Angelo, M. A., Raices, M., Panowski, S. H., Hetzer, M. W. Age-dependent deterioration of nuclear pore complexes causes a loss of nuclear integrity in postmitotic cells. Cell. 136 (2), 284-295 (2009).
  18. Hetzer, M. W. The role of the nuclear pore complex in aging of post-mitotic cells. Aging (Albany NY). 2 (2), 74-75 (2010).
  19. Savas, J. N., Toyama, B. H., Xu, T., Yates, J. R., Hetzer, M. W. Extremely long-lived nuclear pore proteins in the rat brain. Science. 335 (6071), 942 (2012).
  20. Toyama, B. H., et al. Identification of long-lived proteins reveals exceptional stability of essential cellular structures. Cell. 154 (5), 971-982 (2013).
  21. Nofrini, V., Di Giacomo, D., Mecucci, C. Nucleoporin genes in human diseases. European Journal of Human Genetics. 24 (10), 1388-1395 (2016).
  22. Sakuma, S., D’Angelo, M. A. The roles of the nuclear pore complex in cellular dysfunction, aging and disease. Seminars in Cell & Developmental Biology. 68, 72-84 (2017).
  23. Basel-Vanagaite, L., et al. Mutated nup62 causes autosomal recessive infantile bilateral striatal necrosis. Annals of Neurology. 60 (2), 214-222 (2006).
  24. Nousiainen, H. O., et al. Mutations in mRNA export mediator GLE1 result in a fetal motoneuron disease. Nature Genetics. 40 (2), 155-157 (2008).
  25. Cronshaw, J. M., Matunis, M. J. The nuclear pore complex: disease associations and functional correlations. Trends in Endocrinology and Metabolism. 15 (1), 34-39 (2004).
  26. Chou, C. C., et al. TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nature Neuroscience. 21 (2), 228-239 (2018).
  27. Eftekharzadeh, B., et al. Tau protein disrupts nucleocytoplasmic transport in Alzheimer’s disease. Neuron. 99 (5), 925-940 (2018).
  28. Freibaum, B. D., et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature. 525 (7567), 129-133 (2015).
  29. Gasset-Rosa, F., et al. Polyglutamine-expanded huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport. Neuron. 94 (1), 48-57 (2017).
  30. Grima, J. C., et al. Mutant huntingtin disrupts the nuclear pore complex. Neuron. 94 (1), 93-107 (2017).
  31. Jovicic, A., et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nature Neuroscience. 18 (9), 1226-1229 (2015).
  32. Guo, L., et al. Nuclear-import receptors reverse aberrant phase transitions of RNA-binding proteins with prion-like domains. Cell. 173 (3), 677-692 (2018).
  33. Hofweber, M., et al. Phase Separation of FUS Is Suppressed by Its Nuclear Import Receptor and Arginine Methylation. Cell. 173 (3), 706-719 (2018).
  34. Yoshizawa, T., et al. Nuclear Import Receptor Inhibits Phase Separation of FUS through Binding to Multiple Sites. Cell. 173 (3), 693-705 (2018).
  35. Chew, J., et al. Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy. Molecular Neurodegeneration. 14 (1), 9 (2019).
  36. Zhang, Y. J., et al. Poly(GR) impairs protein translation and stress granule dynamics in C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. Nat Medicine. 24 (8), 1136-1142 (2018).
  37. Zhang, Y. J., et al. C9ORF72 poly(GA) aggregates sequester and impair HR23 and nucleocytoplasmic transport proteins. Nature Neuroscience. 19 (5), 668-677 (2016).
  38. Zhang, Y. J., et al. Heterochromatin anomalies and double-stranded RNA accumulation underlie C9orf72 poly(PR) toxicity. Science. 363 (6428), (2019).
  39. Beck, M., Hurt, E. The nuclear pore complex: understanding its function through structural insight. Nature Reviews. Molecular Cell Biology. 18 (2), 73-89 (2017).
  40. Lin, D. H., Hoelz, A. The structure of the nuclear pore complex (an update). Annual Review of Biochemistry. 88, 725-783 (2019).
  41. Raices, M., D’Angelo, M. A. Nuclear pore complex composition: a new regulator of tissue-specific and developmental functions. Nature Reviews. Molecular Cell Biology. 13 (11), 687-699 (2012).
  42. Kinoshita, Y., Kalir, T., Dottino, P., Kohtz, D. S. Nuclear distributions of NUP62 and NUP214 suggest architectural diversity and spatial patterning among nuclear pore complexes. PLoS One. 7 (4), 36137 (2012).
  43. Ori, A., et al. Cell type-specific nuclear pores: a case in point for context-dependent stoichiometry of molecular machines. Molecular Systems Biology. 9, 648 (2013).
  44. Rajoo, S., Vallotton, P., Onischenko, E., Weis, K. Stoichiometry and compositional plasticity of the yeast nuclear pore complex revealed by quantitative fluorescence microscopy. Proceedings of the National Academy of Sciences of the United States of America. 115 (17), 3969-3977 (2018).
  45. Li, C., Goryaynov, A., Yang, W. The selective permeability barrier in the nuclear pore complex. Nucleus. 7 (5), 430-446 (2016).
  46. Raices, M., D’Angelo, M. A. Nuclear pore complexes and regulation of gene expression. Current Opinion in Cell Biology. 46, 26-32 (2017).
  47. Pascual-Garcia, P., Capelson, M. Nuclear pores in genome architecture and enhancer function. Current Opinion in Cell Biology. 58, 126-133 (2019).
  48. Schermelleh, L., et al. Subdiffraction multicolor imaging of the nuclear periphery with 3D structured illumination microscopy. Science. 320 (5881), 1332-1336 (2008).
  49. Maglione, M., Sigrist, S. J. Seeing the forest tree by tree: super-resolution light microscopy meets the neurosciences. Nature Neuroscience. 16 (7), 790-797 (2013).
  50. Thevathasan, J. V., et al. Nuclear pores as versatile reference standards for quantitative superresolution microscopy. Nature Methods. 16 (10), 1045-1053 (2019).
  51. Wu, Y., Shroff, H. Faster, sharper, and deeper: structured illumination microscopy for biological imaging. Nature Methods. 15 (12), 1011-1019 (2018).
  52. Hampoelz, B., et al. Pre-assembled nuclear pores insert into the nuclear envelope during early development. Cell. 166 (3), 664-678 (2016).
  53. Hampoelz, B., et al. Nuclear pores assemble from nucleoporin condensates during oogenesis. Cell. 179 (3), 671-686 (2019).
  54. Agote-Aran, A., et al. Spatial control of nucleoporin condensation by fragile X-related proteins. The EMBO Journal. 39 (20), 104467 (2020).
  55. Colombi, P., Webster, B. M., Fröhlich, F., Lusk, C. P. The transmission of nuclear pore complexes to daughter cells requires a cytoplasmic pool of Nsp1. The Journal of Cell Biology. 203 (2), 215-232 (2013).
  56. Sivaguru, M., et al. Comparative performance of airyscan and structured illumination superresolution microscopy in the study of the surface texture and 3D shape of pollen. Microscopy Research and Technique. 81 (2), 101-114 (2018).
  57. Löschberger, A., Franke, C., Krohne, G., van de Linde, S., Sauer, M. Correlative super-resolution fluorescence and electron microscopy of the nuclear pore complex with molecular resolution. Journal of Cell Science. 127, 4351-4355 (2014).
  58. Löschberger, A., et al. Super-resolution imaging visualizes the eightfold symmetry of gp210 proteins around the nuclear pore complex and resolves the central channel with nanometer resolution. Journal of Cell Science. 125, 570-575 (2012).
This article has been published
Video Coming Soon
Keep me updated:

.

Citar este artigo
Coyne, A. N., Rothstein, J. D. Nuclei Isolation and Super-Resolution Structured Illumination Microscopy for Examining Nucleoporin Alterations in Human Neurodegeneration. J. Vis. Exp. (175), e62789, doi:10.3791/62789 (2021).

View Video