Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Immunology and Infection

Protein Misfolding Cyclic Amplification of Prions

Published: November 7, 2012 doi: 10.3791/4075

Summary

Protein misfolding cyclic amplification (PMCA) is an in vitro assay for the study of prion conversion and strain and species barriers. It can also be used as a prion detection assay.

Abstract

Prions are infectious agents that cause the inevitably fatal transmissible spongiform encephalopathy (TSE) in animals and humans9,18. The prion protein has two distinct isoforms, the non-infectious host-encoded protein (PrPC) and the infectious protein (PrPSc), an abnormally-folded isoform of PrPC 8.

One of the challenges of working with prion agents is the long incubation period prior to the development of clinical signs following host inoculation13. This traditionally mandated long and expensive animal bioassay studies. Furthermore, the biochemical and biophysical properties of PrPSc are poorly characterized due to their unusual conformation and aggregation states.

PrPSc can seed the conversion of PrPC to PrPSc in vitro14. PMCA is an in vitro technique that takes advantage of this ability using sonication and incubation cycles to produce large amounts of PrPSc, at an accelerated rate, from a system containing excess amounts of PrPC and minute amounts of the PrPSc seed19. This technique has proven to effectively recapitulate the species and strain specificity of PrPSc conversion from PrPC, to emulate prion strain interference, and to amplify very low levels of PrPSc from infected tissues, fluids, and environmental samples6,7,16,23 .

This paper details the PMCA protocol, including recommendations for minimizing contamination, generating consistent results, and quantifying those results. We also discuss several PMCA applications, including generation and characterization of infectious prion strains, prion strain interference, and the detection of prions in the environment.

Protocol

1. Preparing the Equipment

  1. Use a Misonix 3000 or Misonix 4000 sonicator (Farmingdale, NY) connected to a Thermo Electron Neslab EX-7 water bath (Newington, NH) to keep a constant temperature of 37 °C. Sonicate the samples in 200 μl thin-walled PCR tube strips with domed caps obtained from Thermo Scientific (Waltham, MA).
  2. A brand new sonicator requires a "break-in" period of continuous operation9. A two-month break-in period consists of a 40-sec sonication burst and 10-minute incubation cycles with the amplitude level set to 10. One should observe erosion throughout the surface of the titanium microplate horn (Figure 1, panel B). The circulating water will look white turbid due to erosion of the titanium microplate horn. Replace this water daily.
  3. The amplification of PrPSc will vary throughout the microplate horn. The best PrPSc amplification efficiency can be obtained within a radius of 5 cm from the center of the microplate horn (See Figure 1, panel C).
  4. One round of PMCA consists 144 cycles. Each cycle consists of 5 sec sonication and 10 min incubation. This will roughly entail to 24 hr per round of PMCA.
  5. The power output of sonication, displayed on the sonicator's control panel, varies with the number of PCR tubes present in the sonicator's rack and the temperature of the circulating water from the water bath. Ensure that the water is equilibrated at 37 °C, and do not fill more than 30% of the sonicator's tube rack (Figure 1, panel C). Spread the PCR tubes through the microplate horn. Have at least one row of empty space between PCR tube strips and allow no more than four tubes connected together per strip (Figure 1, panel C).
  6. The strength of sonication is critical for a successful prion strain amplification. While sonicating, adjust the amplitude to have a power output ranging between 155 ~ 170 watts. In our laboratory the power level is set to six and seven for the Misonix 3000 and Misonix 4000, respectively.
  7. Use distilled water in the water bath to avoid accumulating hard water stains. Replace this water weekly. During the incubation and sonication cycles, water condensation will occur inside the microplate horn lid. To avoid condensation, attach a small aquarium heat pad to the top of the sonicator's enclosure box as shown in Figure 1, panel A.

2. Preparing the Samples

  1. All procedures involving animals were approved by the Creighton University Institutional Animal Care and Use Committee and comply with the Guide for the Care and Use of Laboratory Animals. Before any tissue collection, the animal must be anesthetized and transcardially perfused using ice-cold phosphate buffered saline solution with 5 mM EDTA at pH 7.4 to remove blood from the brain and avoid the inhibition of PMCA reaction by blood7. Collect animal tissues rapidly using aseptic technique and have dedicated dissection tools for each prion strain. After dissection, place the tissue in a 1.5 ml microcentrifuge tube, flash freeze it on dry ice, and store it at -80 °C until homogenization.
  2. To prepare the PMCA substrate (PrPC) solution, homogenize an uninfected hamster brain (UN BH) to a 10% weight/volume (w/v) solution with ice cold conversion buffer (see part 3) using a Tenbroeck tissue grinder. While homogenizing, be careful not to get air inside the grinder to avoid excessive foaming of the solution. Clarify the solution by centrifuging at 500 x g for 30 sec and aliquot the supernatant in 1.5 ml microcentrifuge tubes. Store at -80 °C until further use.
  3. To prepare the PMCA seed (PrPSc) solution, homogenize an infectious material (brain, spleen, lymph nodes, other tissues or contaminated soil) to a 10% w/v solution with ice cold water and aliquot the solution in 0.6 ml microcentrifuge tubes. Store at -80 °C until further use.

3. Preparing Conversion Buffer

  1. Weigh 0.5093 g of Triton X-100 in a 50 ml volumetric flask (result is a 1% final concentration of Triton X-100) (Sigma-Aldrich GmbH; Steinheim, Germany).
  2. Add one Complete Protease Inhibitor Tablet (Roche Diagnostics GmbH; Mannheim, Germany).
  3. Add 0.0931 g of EDTA (this will result in a 5 mM final concentration of EDTA) (Mallinckrodt Baker Inc.; Phillipsburg, NJ).
  4. Adjust the pH to 7.4 with 1N NaOH.
  5. Adjust volume to 50 ml with Dulbecco's Phosphate Buffered Saline (DPBS) (Mediatech Inc.; Manassas, VA).
  6. Filter the solution through a 0.45 μm nitrocellulose membrane with a vacuum filtration apparatus. The conversion buffer can be stored at 4 °C for no more than one month.

4. Amplification of Prion Strains Using PMCA

  1. Dilute the PMCA seed into the PMCA substrate at a ratio of 1:20 (i.e. mix 5 μl of the PMCA seed and 95 μl of the PMCA substrate) into a PCR tube. Remove and store a 15 μl aliquot at -80 °C for unsonicated control; this unsonicated control will be used to quantify the PrPSc amplification via proteinase K (PK) digestion followed by Western blot (WB) analysis. An additional unsonicated control could be generated by placing a second 15 μl aliquot at 37 °C for the duration of the PMCA reaction. A minimum of three repetitions is required for statistical purposes.
  2. Subject the remainder of the sample (70 μl) to one round of PMCA as depicted on Figure 1, Panel A. Shake tubes every 5 hr to avoid water condensation on the domed cap of the PCR tube.
  3. The sample's tubes should be spun down and gently vortexed (being careful that the solution do not go into the domed cap) before opening after any procedure to ensure homogeneity and avoid potential contamination.
  4. For serial PMCA (sPMCA) of short incubation period strains (i.e. HY TME, HaCWD, or 263K), dilute the sonicated material at a ratio of 1:20 by transferring 5 μl of the sonicated sample from round one into 95 μl of fresh uninfected brain homogenate (Figure 2, Panel B).
  5. For sPMCA of long incubation period strains (i.e. DY TME, 139H, 22CH, 22AH, or ME7H), dilute the sonicated material at a ratio of 1:1 by transferring 50 μl of the sonicated sample from round one into 50 μl of fresh uninfected brain homogenate (Figure 2, Panel B).
  6. Remove two 15 μl aliquots from the previously diluted sonicated mixture (from steps 4.4 or 4.5) and store one of them at -80 °C and the other at 37 °C (unsonicated control solutions for PMCA round two). Subject the remnants to a PMCA round two.
  7. This procedure can be repeated indefinitely. In our laboratory, we have performed up to fifteen PMCA rounds, with the strain properties remaining unchanged.
  8. PK digest the samples. For a typical WB, mix 5 μl of PK 80 μg/ml to 5 μl of sample (this solution will have a final PK concentration of 40 μg/ml) and incubate at 37 °C for one hour. Add 10 μl of gel loading buffer. Boil this solution for 10 min. Let the solution cool down and load 10 μl into the gel.
  9. To calculate the success of amplification perform a WB analysis on the PK-digested samples to estimate the amount of amplified PrPSc by comparing them to either, the unsonicated controls20,21 Figure 2, Panel C or to known amounts of PK-undigested recombinant PrP.

5. Avoiding Cross-contamination

Critical steps have to be taken to minimize cross-contamination and the occurrence of false positives due to de novo formation of protease resistant products.

  1. Use a dedicated set of dissection tools (i.e. forceps, tweezers, scissors) for sample collection for each prion strain.
  2. Before homogenizing uninfected brains to be used as a PMCA substrate, wipe the pipettes with 1% bleach. Soak the sonicator's PCR tube rack, tissue homogenizers, and PCR tube storage racks with 1% bleach for 10 min and rinse thoroughly with distilled water.
  3. Cover the work area with a fresh Versi-Dry Lab Soaker every time a new sample is to be homogenized, a new PMCA experiment is to be prepared, or when aliquoting between sPMCA rounds.
  4. Use new gloves each time new samples are handled (especially when transferring aliquots between sPMCA rounds).
  5. Use short sonication bursts for each PMCA cycle. Cycles of 5 sec sonication combined with 10 min incubation is sufficient to amplify prions without generating de novo PrPSc 1,20,21,23,24 . As other laboratories have shown, increasing the sonication time, increasing the sonication amplitude, and extending the number of PMCA cycles increases the probability for de novo PrPSc formation2.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Protein misfolding cyclic amplification (PMCA) is used to amplify PrPSc in vitro7, 12, 14, 19, 24. A successful PrPSc amplification is shown by an increase in band intensity on Western blots of the PK-resistant prion protein (migrating between 19 and 30 kDa for hamster-derived prion strains) as shown in Figure 3. The increase in its band intensity after PMCA indicates amplification of the PK-resistant PrPSc material. Successful amplification of the hamster-derived prion protein, HaCWD and DY TME, is shown in the WB analysis of Figure 3 by comparing the band intensities before (lanes 5 and 7) and after (lanes 4 and 6) PMCA.

PK digestion of PrPSc followed by WB analysis shows an upper, middle and lower band corresponding to the di-, mono-, and unglycosylated prion protein, respectively. Some prion strains can be differentiated by their electrophoretic mobility. For instance, there is a two-kDa difference in migration between the HaCWD and DY TME prion strains. (Figure 3, lanes 1 and 2, respectively).

A high fidelity PrPSc amplification is achieved by the PMCA1, 7, 12, 19, 23, 24. This high fidelity in PMCA amplification is observed by a similar electrophoretic migration of the PMCA-amplified prion strains (HaCWD and DY TME) when compared to their corresponding seeds (Figure 3; lanes 1 & 4 and 2 & 6 for HaCWD and DY TME, respectively).

If no cross-contamination or de novo PrPSc formation occurs, WB analysis of mock control PMCA samples should remain clear after PK digestion (Figure 3, lanes 8 and 9).

Figure 1
Figure 1. To avoid water condensation, a regular aquarium heat-pad is placed above the lid of the microplate horn (panel A). A brand new sonicator should be allowed a break-in period of continuous sonication for about two months. Panel B shows the erosion on the titanium microplate horn after the break-in period. Panel C shows a possible arrangement of PCR tube strips that would allow an optimal sonication of the samples.

Figure 2
Figure 2. Flow chart for PMCA (panel A), sPMCA (panel B), and WB analysis (panel C). PrPSc is the PMCA seed and the uninfected brain homogenate (UN BH) is the PMCA substrate. The quantification of the amplified PrPSc for each PMCA round is obtained by dividing the band intensity after PMCA by its corresponding band intensity before PMCA.

Figure 3
Figure 3. Western blot of PK-digested HaCWD and DY TME hamster-derived prion strains. PrPSc is detected using 3F4 anti-prion antibody. Analysis of the WB shows the abundance (blot intensity) and electrophoretic mobility of PrPSc. PMCA reactions were seeded with brain homogenate from hamsters infected with either the HaCWD (lanes 4-5) or DY TME (lanes 6-7) agents. Samples that underwent PMCA (PMCA +) show an increase in the PrPSc abundance when compared to their unamplified (PMCA -) controls (compare lanes 5 to 4 and 7 to 6). There is a two-kDa difference in the electrophoretic migration between the prion proteins of HaCWD and DY TME (lanes 1-2). This difference in migration is maintained in the PMCA-generated samples using HaCWD and DY TME homogenates (lanes 4 and 6, respectively). PMCA reactions seeded with uninfected brain (mock) homogenate fail to amplify PrPSc (lane 8). The locations for the 19 and 21 kDa molecular markers are indicated to the left of the panel.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Challenges of amplifying infectious prion proteins are the long incubation periods and the expenses of in vivo experiments. The PMCA technique is a cost effective means to amplify infectious prion agents. Several laboratories have confirmed the ability of PMCA to accurately amplify prion strains in vitro 7, 9, 12, 14, 19,24 .

Prion diseases can be transmitted between species. Bessen and Marsh have effectively inoculated hamsters with transmissible mink encephalopathy, which produced two distinct hamster-derived prion strains5 . In an elegant study, Telling and co-workers used sPMCA to amplify the mouse-derived prion strain, RML, using transgenic mice expressing cervid PrPC (Tg(CerPrP)1536+/-) as PMCA substrate. A rapid onset of disease was observed after inoculation of Tg(CerPrP)1536+/- with the PMCA-adapted material12 . Similarly, Kurt et al. were able to amplify chronic wasting disease (CWD), a prion disease in cervids, using non-cervid species PMCA substrate15. These data suggest that species barrier in prion diseases can be bypassed in vitro via PMCA.

Bessen and Marsh have shown that the hamster's short incubation period strain, HY TME, has a quicker PrPSc accumulation when compared to its long incubation period counterpart, DY TME3, 5 . Similarly, this correlation between the incubation period and the accumulation rate is observed after PMCA in several hamster-derived prion strains1, 23, 24.

The amplification rate is a property intrinsic to each strain. PMCA has been used to quantify this amplification rate. Ayers et al. were able to calculate a unitless number, termed amplification coefficient, that represents the rate of amplification for a given hamster-derived prion strain1 .

Prion strains can interfere with each other when present in the same host. The presence of a long incubation period strain can extend the incubation period or even block the ability of a short incubation period strain to cause disease. This extension in incubation period is termed strain interference. Strain interference has been shown to occur in mice10 and hamsters22. Bartz and co-workers have used PMCA to study prion strain interference in hamsters. Their results show that the PMCA experiments correlate with the findings of a similar experiment in vivo22, 23.

Since there is a relatively low level of PrPSc outside the central nervous system, prions can only be accurately diagnosed post-mortem via dissection of the brain followed by immunohistochemistry. PMCA can be used as diagnostic tool, since it has shown a great capability of amplifying minute amounts of PrPSc 7 , even from hamster's urine samples11.

The prion agent is able to withstand harsh environmental conditions and remain infectious16,17 . However, the amount of PrPSc in the environment is too small to be detected using conventional techniques such WB. PMCA has been used to amplify and calculate the approximate amount of PrPSc the environment, such as soil and water16, 17, 20, 21 .

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

No conflicts of interest declared.

Acknowledgments

We would like to thank Dr. Vesper Fe Marie Ramos for critical reading of the manuscript. This work was supported by the National Center for Research Resources (P20 RR0115635-6, C06 RR17417-01 and G20RR024001) and the National Institute for Neurological Disorders and Stroke (2R01 NS052609).

Materials

Name Company Catalog Number Comments
Misonix 3000 Misonix S-3000
Misonix 4000 Misonix S-4000
Tenbr–ck Tissue Grinder Kontes 885000-0007
Neslab EX-7 Water Bath Thermo Electron Neslab EX-7
0.2 ml PCR Tube Strips Thermo Scientific AB-0451
Triton X-100 Sigma Aldrich T9284-100ML
Complete Protease Inhibitor Roche 11 697 498 001
EDTA J.T. Baker 4040-00
DPBS Mallinckrodt Baker Mediatech 21-031-CV
Versi-Dry Lab Soakers Fisher Scientific 14 206 28
Repti Therm Heater Zoo Med Laboratories, Inc. RH-4

DOWNLOAD MATERIALS LIST

References

  1. Ayers, J. I., Schutt, C. R., Shikiya, R. A., Aguzzi, A., Kincaid, A. E., Bartz, J. C. The strain-encoded relationship between PrP replication, stability and processing in neurons is predictive of the incubation period of disease. PLoS pathogens. 7, e1001317 (2011).
  2. Barria, M. A., Mukherjee, A., Gonzalez-Romero, D., Morales, R., Soto, C. De novo generation of infectious prions in vitro produces a new disease phenotype. PLoS Pathog. 5, e1000421 (2009).
  3. Bessen, R. A., Marsh, R. F. Biochemical and physical properties of the prion protein from two strains of the transmissible mink encephalopathy agent. J. Virol. 66, 2096-2101 (1992).
  4. Bessen, R. A., Marsh, R. F. Distinct PrP properties suggest the molecular basis of strain variation in transmissible mink encephalopathy. J. Virol. 68, 7859-7868 (1994).
  5. Bessen, R. A., Marsh, R. F. Identification of two biologically distinct strains of transmissible mink encephalopathy in hamsters. J. Gen. Virol. 73, 329-334 (1992).
  6. Castilla, J., Gonzalez-Romero, D., Saa, P., Morales, R., De Castro, J., Soto, C. Crossing the species barrier by PrP(Sc) replication in vitro generates unique infectious prions. Cell. 134, 757-768 (2008).
  7. Castilla, J., Morales, R., Saa, P., Barria, M., Gambetti, P., Soto, C. Cell-free propagation of prion strains. EMBO J. 27, 2557-2566 (2008).
  8. Caughey, B., Raymond, G. J. The scrapie-associated form of PrP is made from a cell surface precursor that is both protease- and phospholipase-sensitive. J. Biol. Chem. 266, 18217-18223 (1991).
  9. Deleault, N. R., Harris, B. T., Rees, J. R., Supattapone, S. Formation of native prions from minimal components in vitro. Proceedings of the National Academy of Sciences of the United States of America. 104, 9741-9746 (2007).
  10. Dickinson, A. G., Fraser, H., Meikle, V. M., Outram, G. W. Competition between different scrapie agents in mice. Nat. New Biol. 237, 244-245 (1972).
  11. Gonzalez-Romero, D., Barria, M. A., Leon, P., Morales, R., Soto, C. Detection of infectious prions in urine. FEBS Lett. 582, 3161-3166 (2008).
  12. Green, K. M., Castilla, J., Seward, T. S., Napier, D. L., Jewell, J. E., Soto, C., Telling, G. C. Accelerated high fidelity prion amplification within and across prion species barriers. PLoS Pathog. 4, e1000139 (2008).
  13. Hadlow, W. J., Race, R. E., Kennedy, R. C. Temporal distribution of transmissible mink encephalopathy virus in mink inoculated subcutaneously. J. Virol. 61, 3235-3240 (1987).
  14. Kocisko, D. A., Come, J. H., Priola, S. A., Chesebro, B., Raymond, G. J., Lansbury, P. T., Caughey, B. Cell-free formation of protease-resistant prion protein. Nature. 370, 471-474 (1994).
  15. Kurt, T. D., Telling, G. C., Zabel, M. D., Hoover, E. A. Trans-species amplification of PrP(CWD) and correlation with rigid loop 170N. Virology. 387, 3235-3240 (2009).
  16. Maddison, B. C., Baker, C. A., Terry, L. A., Bellworthy, S. J., Thorne, L., Rees, H. C., Gough, K. C. Environmental sources of scrapie prions. J. Virol. 84, 11560-11562 (2010).
  17. Nichols, T. A., Pulford, B., Wyckoff, A. C., Meyerett, C., Michel, B., Gertig, K., Hoover, E. A., Jewell, J. E., Telling, G. C., Zabel, M. D. Detection of protease-resistant cervid prion protein in water from a CWD-endemic area. Prion. 3, 171-183 (2009).
  18. Prusiner, S. B. Novel proteinaceous infectious particles cause scrapie. Science. 216, 136-144 (1982).
  19. Saborio, G. P., Permanne, B., Soto, C. Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding. Nature. 411, 810-813 (2001).
  20. Saunders, S. E., Bartz, J. C., Vercauteren, K. C., Bartelt-Hunt, S. L. An enzymatic treatment of soil-bound prions effectively inhibits replication. Appl. Environ. Microbiol. 77, 4313-4317 (2011).
  21. Saunders, S. E., Shikiya, R. A., Langenfeld, K., Bartelt-Hunt, S. L., Bartz, J. C. Replication efficiency of soil-bound prions varies with soil type. Journal of virology. , (2011).
  22. Schutt, C. R., Bartz, J. C. Prion interference with multiple prion isolates. Prion. 2, 61-63 (2008).
  23. Shikiya, R. A., Ayers, J. I., Schutt, C. R., Kincaid, A. E., Bartz, J. C. Co-infecting prion strains compete for a limiting cellular resource. Journal of. 84, 5706-5714 (2010).
  24. Shikiya, R. A., Bartz, J. C. In vitro generation of high titer prions. Journal of virology. , (2011).
  25. Weber, P., Giese, A., Piening, N., Mitteregger, G., Thomzig, A., Beekes, M., Kretzschmar, H. A. Generation of genuine prion infectivity by serial PMCA. Veterinary microbiology. 123, 346-357 (2007).

Tags

Protein Misfolding Cyclic Amplification Prions Transmissible Spongiform Encephalopathy PrPC PrPSc Incubation Period Clinical Signs Animal Bioassay Studies Biochemical Properties Biophysical Properties In Vitro Technique Sonication Incubation Cycles PrPSc Seed Species Specificity Strain Specificity Prion Strain Interference Amplification Infected Tissues Fluids Environmental Samples PMCA Protocol Contamination Control Consistent Results Quantification
Protein Misfolding Cyclic Amplification of Prions
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Saunders, S. E., Bartz, J. C.,More

Saunders, S. E., Bartz, J. C., Shikiya, R. A. Protein Misfolding Cyclic Amplification of Prions. J. Vis. Exp. (69), e4075, doi:10.3791/4075 (2012).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter