Waiting
Elaborazione accesso...

Trial ends in Request Full Access Tell Your Colleague About Jove

Neuroscience

Generation of Human Brain Organoids for Mitochondrial Disease Modeling

Published: June 21, 2021 doi: 10.3791/62756

Summary

We describe a detailed protocol for the generation of human induced pluripotent stem cell-derived brain organoids and their use in modeling mitochondrial diseases.

Abstract

Mitochondrial diseases represent the largest class of inborn errors of metabolism and are currently incurable. These diseases cause neurodevelopmental defects whose underlying mechanisms remain to be elucidated. A major roadblock is the lack of effective models recapitulating the early-onset neuronal impairment seen in the patients. Advances in the technology of induced pluripotent stem cells (iPSCs) enable the generation of three-dimensional (3D) brain organoids that can be used to investigate the impact of diseases on the development and organization of the nervous system. Researchers, including these authors, have recently introduced human brain organoids to model mitochondrial disorders. This paper reports a detailed protocol for the robust generation of human iPSC-derived brain organoids and their use in mitochondrial bioenergetic profiling and imaging analyses. These experiments will allow the use of brain organoids to investigate metabolic and developmental dysfunctions and may provide crucial information to dissect the neuronal pathology of mitochondrial diseases.

Introduction

Mitochondrial diseases represent the largest class of inborn errors of metabolism1. They are caused by genetic mutations disrupting different mitochondrial processes, including oxidative phosphorylation (OXPHOS)2, respiratory chain assembly, mitochondrial dynamics, and mitochondrial DNA transcription or replication3. Tissues with energy requirements are particularly affected by mitochondrial dysfunction4. Accordingly, patients with mitochondrial diseases typically develop early-onset neurological manifestations.

There are currently no treatments available for children affected with mitochondrial diseases5. A major hindrance for drug development of mitochondrial diseases is the lack of effective models recapitulating the human disease course6. Several of the currently studied animal models do not exhibit the neurological defects present in the patients7. Hence, the mechanisms underlying the neuronal pathology of mitochondrial diseases are still not fully understood.

Recent studies generated iPSCs from patients affected by mitochondrial diseases and used these cells to obtain patient-specific neuronal cells. For example, genetic defects associated with the mitochondrial disease, Leigh syndrome, have been found to cause aberrations in cellular bioenergetics8,9, protein synthesis10, and calcium homeostasis9,11. These reports provided important mechanistic clues on the neuronal impairment occurring in mitochondrial diseases, paving the way for drug discovery for these incurable diseases12.

Two-dimensional (2D) cultures, however, do not enable the investigation of the architectural complexity and regional organization of 3D organs13. To this end, the use of 3D brain organoids derived from patient-specific iPSCs14 may allow researchers to gain additional important information and thereby help to dissect how mitochondrial diseases impact the development and function of the nervous system15. Studies employing iPSC-derived brain organoids to investigate mitochondrial diseases are beginning to uncover the neurodevelopmental components of mitochondrial diseases.

Spinal cord organoids carrying mutations associated with the mitochondrial disease, mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes syndrome (MELAS), showed defective neurogenesis and delayed motor neuron differentiation16. Cortical organoids derived from patients with the mitochondrial disease, Leigh syndrome, showed reduced size, defects in neural epithelial bud generation, and loss of cortical architecture17. Brain organoids from Leigh syndrome patients showed that the disease defects initiate at the level of neural progenitor cells, which cannot commit to mitochondrial metabolism, causing aberrant neuronal branching and morphogenesis18. Thus, neural progenitors may represent a cellular therapeutic target for mitochondrial diseases, and strategies promoting their mitochondrial function may support the functional development of the nervous system.

The use of brain organoids might help uncover the neurodevelopmental components of mitochondrial diseases. Mitochondrial diseases are mainly considered as early-onset neurodegeneration5. However, neurodevelopmental defects are also present in patients affected by mitochondrial diseases, including developmental delay and cognitive impairment19. Patient-specific brain organoids may help address these aspects and elucidate how mitochondrial diseases may impact human brain development. Mitochondrial dysfunction could also play a pathogenetic role in other more common neurological diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease4. Hence, elucidating the impact of mitochondrial defects in neurodevelopment using brain organoids might also be instrumental for the study of those diseases. This paper describes a detailed protocol for generating reproducible brain organoids that can be used for conducting disease modeling of mitochondrial diseases.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

NOTE: The use of human iPSCs may require an ethical approval. iPSCs used in this study were derived from healthy control individuals following local ethical approval (#2019-681). All cell culture procedures must be performed under a sterile cell culture hood, carefully disinfecting all reagents and consumables before transferring under the hood. Human iPSCs used for differentiation should have a passage number below 50 to avoid potential genomic aberrations that may occur upon extensive culture. The pluripotent state of the cells should be validated before organoid generation, for example, by monitoring the expression of pluripotency-associated markers such as NANOG or OCT4. Mycoplasma tests should be conducted weekly to ensure mycoplasma-free cultures.

1. Generation of brain organoids

  1. Culture of human iPSCs
    1. Culture human iPSCs under feeder-free conditions in iPSC medium (see the Table of Materials) on coated 6-well plates and keep them in a humidified tissue culture incubator at 37 °C and 5% CO2.
      ​NOTE: The carryover of feeder cells may hamper the organoid differentiation. Passage the cells at least once in feeder-free conditions.
    2. Passage the iPSCs at 80% confluency using enzyme-free detachment medium in ratios ranging from 1:4 to 1:12. To increase cell survival, add 10 µM Rho-associated protein kinase (ROCK) inhibitor (Y27632) after each splitting.
  2. Dissociate the iPSCs (80% confluency)-Day 0.
    1. Prepare Cortical Differentiation Medium I (CDMI) (Table 1). Prewarm CDMI medium at room temperature (22-25 °C) before adding it to the cells.
    2. Wash the wells containing the iPSCs with phosphate-buffered saline (PBS) to remove dead cells and debris.
    3. Add 500 µL of prewarmed Reagent A (Table of Materials) to each well and incubate for 5 min at 37 °C. Check under the microscope to ensure cell detachment.
    4. Add 1 mL of iPSC medium to dilute reagent A to neutralize its activity.
    5. Use a 1000 µL pipette to dissociate the cells by pipetting up and down and transfer the cell suspension to a 15 mL centrifuge tube.
    6. Gently centrifuge the iPSCs at 125 x g for 5 min at room temperature (22-25 °C).
    7. Carefully aspirate the supernatant to avoid disturbing the cell pellet.
    8. Resuspend the pellet with 1 mL of CDMI to obtain a single-cell suspension, and count the cell number.
    9. Prepare the seeding medium with 9,000 iPSCs per 100 µL in CDMI supplemented with 20 µM ROCK inhibitor, 3 µM WNT-catenin inhibitor (IWR1), and 5 µM SB431542.
    10. Add 100 µL of seeding medium per well to a 96-well v-bottom plate.
    11. Keep the plate in a humidified tissue culture incubator at 37 °C and 5% CO2.
  3. Neurosphere generation
    1. On day 1, observe that round cell aggregates (neurospheres) with defined smooth borders are forming. Note the dead cells around the aggregates. Continue to culture in the incubator at 37 °C and 5% CO2.
    2. On day 3, agitate the plate by tapping on the sides three times to detach dead cells.
    3. Add 100 µL of CDMI supplemented with 20 µM ROCK inhibitor, 3 µM IWR1, and 5 µM SB431542 to each well.
    4. Return the plate to the incubator at 37 °C and 5% CO2.
    5. On day 6, carefully remove 80 µL of the supernatant medium from each well. Avoid touching the bottom of the well.
    6. Add 100 µL of CDMI supplemented with 3 µM IWR1 and 5 µM SB431542 to each well. Return the plate to the incubator at 37 °C and 5% CO2.
    7. Repeat steps 5 and 6 every 3 days until day 18.
  4. Transfer of neurospheres
    1. On day 18, prepare Cortical Differentiation Medium II (CDMII) (Table 1) and add 10 mL to a 100 mm ultra-low attachment cell culture plate.
    2. Use a 200 µL pipette with the tip cut off to transfer the round neurospheres from the 96-well plate to the 100 mm ultra-low attachment cell culture plate.
      NOTE: Be gentle to avoid damaging the neurospheres by making sure the opening of the tip is wide enough and that the aggregates are not aspirated too quickly.
    3. Remove 5 mL of medium from the plate containing the neurospheres and add 5 mL of fresh CDMII.
      NOTE: This procedure helps to reduce the amount of CDMI medium that may have carried over from the transfer of neurospheres.
    4. Place the plate on an orbital shaker at 70 rpm inside a humidified tissue culture incubator at 37 °C and 5% CO2.
      NOTE: Visually inspect the neurospheres the next day. Increase the speed of the orbital shaker if the neurospheres are clumped together or attached to the bottom of the plate.
    5. Every 3 days, carefully aspirate the supernatant medium and replace it with fresh CDMII. Leave a small amount of the medium to prevent the neurospheres from drying out.
    6. On day 35, prepare Cortical Differentiation Medium III (CDMIII) (Table 1).
      NOTE: The matrix component should be dissolved in cold CDMIII.
    7. Aspirate the medium from the plate and add 10 mL of cold CDMIII.
      NOTE: It is more effective to use cold medium so that the matrix component can coat the organoids without forming clumps.
    8. After changing the medium, place the plate back on an orbital shaker at 70 rpm inside a humidified tissue culture incubator at 37 °C and 5% CO2.
    9. Change the medium every 3-5 days depending on the rate of growth, as indicated by the color of the medium.
    10. On day 70, prepare Cortical Differentiation Medium IV (CDMIV) (Table 1). Use CDMIV medium until the desired age of organoids is reached. During this period, keep the plate on an orbital shaker set at 70 rpm inside a humidified tissue culture incubator (37 °C and 5% CO2).
    11. Change the medium every 3-5 days, depending on the growth rate.

2. Immunostaining of brain organoids

  1. Tissue preparation
    1. Prepare 4% paraformaldehyde (PFA) solution, and place it under a safety hood.
      NOTE: Wear personal safety equipment when handling PFA.
    2. Collect brain organoids and gently transfer them with a blunt-tipped 3 mL plastic Pasteur pipette to a 6-well plate filled with PFA.
      NOTE: Use organoids older than 40 days to allow the visualization of structures with higher cellular complexity.
    3. Keep the organoids in the PFA solution for 1 h at room temperature.
    4. Carefully remove the PFA with a 3 mL plastic Pasteur pipette, and wash the fixed organoids three times using PBS.
    5. Store the fixed organoids at 4 °C in PBS until further use.
  2. Preparation of brain organoid slices
    1. Prepare a 3% agar solution and heat slowly until liquefied.
    2. Place the mold (the cut end of a 10 mL syringe) on a piece of absorbent filter paper (smooth side up). Place a droplet of agar onto it.
    3. Quickly take a single organoid out of the 6-well plate with a spatula and remove excessive PBS with filter paper.
      NOTE: Be careful not to touch the organoid directly with filter paper.
    4. Place the organoid onto the agar droplet.
    5. Repeat this procedure with up to three organoids.
      NOTE: Work fast to avoid solidification of the agar during this step.
    6. Refill the mold with agar until all the organoids are fully covered.
    7. Wait until the agar begins to solidify, and then gently transfer the entire mold containing the organoids, including the absorbent filter paper, onto a cooling element.
      NOTE: If a cooling element is unavailable, store the organoids for a few minutes in a refrigerator at 4 °C.
    8. In the meantime, prepare for the slicing procedure: place a razor blade (cleaned with acetone and washed with double-distilled water) into the holder of the vibratome, mount on the bath, and fill it with PBS.
    9. Remove the mold from (solidified) agar and use a scalpel to trim it to form a cube.
    10. Attach the agar cube containing the organoids on the carrier plate of the vibratome with adhesive gel (see the Table of Materials), and place it in the bath containing PBS.
    11. Adjust the vibratome (see the Table of Materials) to cut slices at a thickness of 150 µm.
      NOTE: Vibratome settings (proper angle, amplitude, frequency, and velocity of the blade) may be similar to those used for slicing fixed brain tissue derived from early postnatal animals. However, the ideal settings depend strongly on the type of the vibratome and must be determined in a first step to prevent distortion or even ripping of the tissue while cutting.
    12. Start the cutting procedure. Use a glass pipette or a spatula to gently transfer each freshly cut slice into a 24-well plate filled with PBS.
    13. Store the plate containing slices at 4 °C (for up to a few days) until further processing.
    14. Transfer the slices out of the plate with a glass pipette or a spatula onto microscope slides. Use a minimum of 2 slices per slide.
    15. Carefully remove the agar and excess PBS with a syringe.
    16. Allow the slices to dry until they adhere to the slides.
      NOTE: While microscope slides can be stored in plastic chambers filled with PBS at 4 °C, they should be stained as soon as possible after the slicing procedure.
  3. Immunohistochemical staining
    1. Prepare the blocking solution (Table 1).
    2. Use a PAP pen to draw a hydrophobic border around the slices on the slide to help keep all the solutions on the slides.
    3. Carefully add the blocking solution on the slide, and incubate for 1 h at room temperature (22-25 °C). To avoid destroying the tissue, do not add the solution directly on top of the slices.
    4. Aspirate the blocking solution and apply the desired primary antibody diluted in the blocking solution.
    5. Incubate the slide overnight in a humidified chamber at 4 °C.
    6. Rinse the slide three times with 1x PBS for 10 min each.
    7. Incubate the slices with the specific secondary antibody diluted in the blocking solution and perform Hoechst staining (1:2,500) for 1 h at room temperature in the dark.
      NOTE: Remember to perform negative controls to confirm there are no non-specific binding or auto-fluorescence.
    8. Rinse three times with 1x PBS for 10 min each in the dark.
    9. Add one drop of mounting medium to the slice, place a coverslip on the edge of the drop, and slowly lay the coverslip down toward the slice to avoid air bubbles.
    10. Allow the slide to rest overnight at room temperature. Apply nail polish on the border of the coverslip to further seal the slide. For long-term storage, store at 4 °C.
  4. Documentation of staining
    1. To scan large images for stitching, utilize a motorized upright wide-field microscope equipped with (see the Table of Materials for details) a high-quality objective; DAPI filter set (e.g., excitation (EX): 340-380 nm, dichroic mirror (DM): 400 nm, barrier filter (BA): 435-485 nm); fluorescein isothiocyanate filter set (e.g., EX: 465-495 nm, DM: 505 nm, BA: 515-555 nm); Alexa594 filter set (e.g., ET 575/40; T 600 LPXR; HC 623/24); digital camera; high-performance acquisition software allowing for automated stitches and stack operations.
    2. For image handling, use an image processing program capable of generating 8-bit tif-files, cropping stitches, adjusting contrast and brightness, merging the channels (e.g., blue, green, and red), and adding scale bars.
    3. To scan details, use a motorized confocal laser scanning microscope equipped with a high-quality objective, a UV laser (EX: 408 nm), an Argon laser (EX: 488 nm), a Helium-Neon laser (EX: 543), imaging software for a confocal microscope.
    4. For image handling of details, use an image processing program capable of generating maximum-intensity projections of confocal z-stacks (e.g., optical sections of 0.6 µm each), generating 8-bit tif-files, adjusting contrast and brightness, merging the channels (e.g., blue, green, and red), adding scale bars.
    5. Use a graphic editor to arrange the figures.

3. Bioenergetic profiling of brain organoids

  1. Preparation of organoids for bioenergetic profiling
    1. Prepare the papain and DNase solution following the manufacturer's protocol.
    2. Transfer 3-5 organoids into a 6-well plate. Wash them two times with prewarmed PBS.
    3. Add 2 mL of prewarmed activated papain solution containing DNase. Using a blade, cut the organoids into small pieces.
    4. Place the plate onto an orbital shaker set at 27 rpm inside a cell culture incubator (at 37 °C, 5% CO2), and incubate for 15-20 min.
      NOTE: The time of incubation depends on the organoid stage. Early-stage organoids can be used as they are. For organoids older than 3 months, it is recommended to cut the organoids into 2-3 pieces before dissociation and incubate the pieces at a rocking speed set at 27 rpm for 15-20 min at 37 °C. This procedure can help remove necrotic tissue that may be present in the later-stage organoids.
    5. Collect the digested tissues into a 15 mL tube and add 5 mL of organoid culture medium CDMIV (Table 1).
    6. Triturate the tissue with a 10 mL plastic pipette by pipetting up and down 10-15 times. Let the undissociated tissue settle down to the bottom of the tube.
    7. Carefully transfer the cell suspension to a 15 mL tube, avoiding any pieces of undissociated tissue. Filter the solution through a 40 µm cell strainer (e.g., polystyrene round-bottom tubes with cell strainer caps).
    8. Pellet the cells by centrifuging at 300 x g for 5 min at room temperature.
    9. Assess the cell number and quality using trypan blue.
    10. Plate the desired number (~20,000/well) of cells onto coated 96-well microplates. Change the medium 6-8 h after plating to Neuronal Medium (Table 1).
    11. Incubate the coated 96-well microplate in a CO2 incubator (37 °C, 5% CO2) for 4 days.
  2. Bioenergetic profiling
    1. On day 3 after replating the dissociated cells, add 200 µL of calibration solution into each well of the bottom part of the 96-well microplate, and place the top green sensor cartridge onto the hydrated microplate.
      NOTE: Place the sensor cartridge on top of the microplate in the correct orientation, and ensure that the calibrant solution covers all the sensors.
    2. Incubate the hydrated 96-well microplate in a non-CO2 incubator at 37 °C overnight.
    3. Turn on the analyzer to allow the instrument to stabilize at 37 °C overnight.
    4. On day 4 after replating, inspect the disassociated organoid culture on the 96-well microplate under the microscope to ensure that the cells appear as a confluent monolayer.
    5. Prepare Assay Medium (Table 1).
    6. Remove Neuronal Medium from all wells with a pipette without touching the bottom of the well to prevent cell damage. Alternatively, carefully invert the whole plate and then dry it on clean paper. Work quickly to avoid cell death.
    7. Wash the cells twice with prewarmed 200 µL of Assay Medium. Add Assay Medium to a final volume of 180 µL per well. Incubate the 96-well microplate in a non-CO2 incubator at 37 °C for 1 h.
    8. Prepare 10 µM solutions of mitochondrial inhibitors in Assay medium. Note that the final concentration after injection is 1 µM.
    9. Load the sensor cartridge placed in the hydrated microplate with 10x solutions of the mitochondrial inhibitors.
      1. Add 18 µL of mitochondrial inhibitor 1 into port A.
      2. Add 19.8 µL of mitochondrial inhibitor 2 into port B.
      3. Add 21.6 µL of mitochondrial inhibitor 2 into port C.
      4. Add 23.4 µL of mitochondrial inhibitor 3 into port D.
    10. Place the loaded cartridge in the hydrated microplate in a non-CO2 incubator at 37 °C until the start of the assay.
    11. Set up a running protocol in the instrument's software (Table 2).
    12. Press START. Take the loaded cartridge from the non-CO2 incubator and place it into the analyzer for calibration.
      NOTE: Make sure the plate is inserted in the correct orientation and without the lid.
    13. Once the calibration step ends, remove the calibration plate. Take the 96-well microplate from the non-CO2 incubator and place it into the analyzer. Click on CONTINUE to start the measurements.
    14. When the run is finished, remove the 96-well cell culture microplate from the analyzer and collect the medium from all the wells without disturbing the cells.
      NOTE: The medium can be stored at -20 °C and used later for measuring the amount of lactate released by the cells in the medium using an appropriate lactate assay kit.
    15. Wash the cells with 200 µL of 1x PBS in each well.
    16. After removing the PBS, freeze the plate at -20 °C.
      NOTE: The frozen plate can be used to quantify cells, proteins, or DNA in each well of the microplate. This quantification will be needed for normalizing the obtained bioenergetic rates. Follow the manufacturer´s instructions for cell, protein, or DNA quantification assays.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

The protocol described here facilitates the robust generation of round organoids (Figure 1A). The generated organoids contain mature neurons that can be visualized using protein markers specific for axons (SMI312) and dendrites (microtubule-associated protein 2 (MAP2)) (Figure 1B). Mature organoids contain not only neuronal cells (MAP2-positive) but also glial cells (e.g., positive for the astrocyte marker S100 calcium-binding protein B (S100ß)) (Figure 1B).

By analyzing sliced brain organoids using confocal microscopy, it is possible to identify and monitor the detailed distribution and organization of different cell types and cellular structures. This could provide new insight into how mitochondrial diseases might affect nervous system development. For example, it is possible to monitor neuronal axons (SMI312-positive) and dendrites (MAP2-positive) (Figure 2A) or the mutual occurrence of neuronal cells (MAP2-positive) and glial cells (S100ß-positive) (Figure 2A). Confocal images may also help to investigate in more detail the distribution and organization of neural progenitors ((sex determining region Y) box-2 (SOX2)-positive) with respect to neurons (beta-III tubulin (TUJ1)-positive) (Figure 2B). Finally, brain organoids can be stained for mitochondria-specific markers (such as the outer mitochondrial membrane protein, translocase of outer membrane 20 kDa subunit (TOM20)) (Figure 2C).

The described protocol enables researchers to perform bioenergetic profiling of brain organoids. Using this procedure, it is possible to measure both mitochondrial metabolism using the oxygen consumption rate (OCR) (Figure 2D) and the glycolytic metabolism using the extracellular acidification rate (ECAR) (Figure 2E). Bioenergetic profiling allows monitoring how cells may modify their OCR and ECAR profiles in response to a sequential administration of mitochondrial inhibitors.

First, the ATP synthase inhibitor, oligomycin, can be applied. Oligomycin causes a drop in the OCR profile (Figure 2D), and therefore, identifies the OCR needed for ATP production. Upon oligomycin treatment, there may also be a compensatory increase in ECAR (Figure 2E), suggesting that the cells can upregulate glycolysis to prevent the metabolic stress caused by the reduction in mitochondrial metabolism. The subsequent double application of the proton ionophore, carbonyl cyanide-p-trifluoromethoxyphenylhydrazon (FCCP), causes the loss of the mitochondrial membrane potential. As the oxygen molecules are now free to move, this causes a rapid increase in OCR (Figure 2D).

These changes in the OCR profile identify the maximal respiration capacity of the cells. The final administration of rotenone plus antimycin A causes a block of the electron transport, and therefore, a steep decrease in OCR (Figure 2D). ECAR may show fluctuation after treatment with FCCP and rotenone plus antimycin A (Figure 2E), depending on the residual glycolytic capacity of the cells. The OCR and ECAR profiles may be dramatically altered in brain organoids derived from mitochondrial patients.

Figure 1
Figure 1: Generation of brain organoids from human iPSCs. (A) Schematic representation of the protocol used to produce brain organoids with corresponding transmission images. Day 0 corresponds to the dissociation of iPSCs and seeding in a 96-well plate with V-bottom using CDMI supplemented with a ROCK inhibitor, a WNT inhibitor, and SB431542. At day 18, neurospheres are transferred from the 96-well plates to 100 mm cell culture dishes with CDMII supplemented with N2. From this point onwards, the cultures are positioned on an orbital shaker. At day 35, the medium is switched from CDMII to CDMIII, which also contains a dissolved matrix component (Table 1). From day 70 onwards, CDMIII is switched to CDMIV supplemented with B27. A representative neurosphere image was taken at day 12 using a microscope camera with 10x magnification. An early organoid image was taken at day 22 using a microscope camera at 4x magnification. A mature organoid image was taken at day 40 using a microscope camera with 4x magnification. (B) The overall structure and cellular organization of brain organoids can be visualized using wide-field microscopy. Representative stitched wide-field images are shown to visualize the relationships between dendrites (MAP2-positive) and axons (SMI312-positive), and between neuronal cells (MAP2-positive) and presumed astrocytes (S100ß-positive). The cells were counterstained with Hoechst to reveal the nuclei. All images were taken using 78 day-old brain organoids. The right column shows the overlay of three channels (merge). Scale bars = 500 µm. Abbreviations: iPSC = induced pluripotent stem cell; CDM = Cortical Differentiation Medium; ROCK = Rho kinase; MAP2 = microtubule-associated protein 2; S100ß= S100 calcium-binding protein B. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Visualization and bioenergetic profiling of brain organoids for mitochondrial disease modeling. The detailed organization and architecture of organoids can be analyzed using confocal microscopy. All images were taken using 78 day-old brain organoids and counterstained with Hoechst to reveal the nuclei. The right column shows the overlay of three channels (merge). Scale bars = 50 µm. (A) Representative extended-focus projections (44-48 optical planes, 0.6 µm each) addressing the interplay between dendrites (MAP2-positive, arrowheads) and axons (SMI312-positive, arrows), and between neuronal cells (MAP2-positive, arrowheads) and presumed astrocytes (S100ß-positive, arrows). (B) Representative extended-focus projections (14-31 optical planes, 0.6 µm each) showing the distribution of neurons (TUJ1-positive, arrowheads) with respect to neural progenitors (SOX2-positive, arrows). (C) Representative extended-focus projections (20 optical planes, 0.6 µm each) showing the distribution within neurons (TUJ1-positive, arrowheads) of mitochondria (visualized using antibodies against the outer mitochondrial membrane protein TOM20, arrows). (D) Mitochondrial respiration of brain organoids can be monitored based on the profile of the OCR after sequential administration of different mitochondrial inhibitors (see text for details). (E) Glycolytic activity of brain organoids can be monitored based on the ECAR upon sequential administration of mitochondrial inhibitors (see text for details). For bioenergetic profiling, approximately 10-15 brain organoids were dissociated to obtain enough cells for replating onto the 96-well microplate. The bars indicate the SEMs based on the results obtained in two independent experiments. Abbreviations: MAP2 = microtubule-associated protein 2; S100ß = S100 calcium-binding protein B; TUJ1 = beta-III tubulin; SOX2 = (sex determining region Y) box-2; TOM20 = translocase of outer membrane 20 kDa subunit; OCR = oxygen consumption rate; ECAR = extracellular acidification rate; Oligom. = oligomycin; FCCP = carbonyl cyanide-p-trifluoromethoxyphenylhydrazon; R = rotenone; AntA = Antimycin A; SEMs = standard error of means. Please click here to view a larger version of this figure.

Media Composition
CDMI (Day 0-18) Final conc.
Glasgow-MEM Gibco 11710-035 [1:1]
Knockout Serum Replacement (KSR) Gibco 10828010 20%
MEM-NEAA (MEM non-essential amino acid solution) Gibco 11140-050 0.1 mM
Sodium Pyruvate Gibco 11360070 1 mM
2-mercaptethanol Gibco 31350-010 0.1 mM
Penicillin and streptomycin Gibco 15140-122 100 U/mL and 100 μg/mL
CDMII (Day 18-35) Final conc.
DMEM/F12 Gibco 31330038 [1:1]
Glutamax Gibco 35050-061 2 mM
N-2 Supplement (100x) Gibco 17502-048 1%
Chemically Defined Lipid Concentrate Gibco 11905031 1%
Penicillin and streptomycin Gibco 15140-122 100 U/mL and 100 μg/mL
CDMIII (Day 35-70) Final conc.
DMEM/F12 Gibco 31330038 [1:1]
Glutamax Gibco 35050-061 2 mM
N-2 Supplement (100x) Gibco 17502-048 1%
Chemically Defined Lipid Concentrate Gibco 11905031 1%
Penicillin and streptomycin Gibco 15140-122 100 U/mL and 100 μg/mL
Fetal Bovine Serum (FBS) Gibco 10270-106 10%
Heparin Merck H3149-25KU 5 μg/mL
Matrigel Corning 356231 1%
CDMIV(Day 70+) Final conc.
DMEM/F12 Gibco 31330038 [1:1]
Glutamax Gibco 35050-061 2 mM
N-2 Supplement (100x) Gibco 17502-048 1%
Chemically Defined Lipid Concentrate Gibco 11905031 1%
Penicillin and streptomycin Gibco 15140-122 100 U/mL and 100 μg/mL
Fetal Bovine Serum (FBS) Gibco 10270-106 10%
Heparin Merck H3149-25KU 5 μg/mL
Matrigel Corning 356231 2%
B-27 supplement with Vitamin A 50x Gibco 17504044 2%
Neuronal Medium Final conc.
DMEM/F12 Gibco 31330038 [1:1]
N-2 supplement Gibco 17502048 [1x]
B-27 supplement with vitamin A 50x Gibco 17504044 [1x]
L-Ascorbic acid Sigma Aldrich A92902 [200 µM]
db-cAMP (dibutyryl cyclic adenosine monophosphate) Sigma Aldrich D0627 500 µM
BDNF (brain-derived neutrotrophic factor) MACS Miltenyi 130-093-811 [10 ng/mL]
GDNF (glial cell line-derived neurotrophic factor) MACS Miltenyi 130-096-290 [10 ng/mL]
Human TGF-β3 (transforming growth factor-beta3) MACS Miltenyi 130-094-007 [1 ng/mL]
Assay Medium Final conc.
Seahorse XF DMEM medium Seahorse Bioscience 103680-100 500 mL
Sodium Pyruvate Gibco 11360070 1 mM
L-Glutamine Lonza BEBP17-605E 2 mM
Glucose Sigma Aldrich 50-99-7 10 mM
Blocking Solution Final conc.
PBS-Tween [1:1] 0.1% Tween
Donkey Serum Sigma Aldrich D9663 10%
Triton-X Merck X100-5ML 1%

Table 1: Details of media and solutions used for organoid generation.

Initialization Baseline (X3) Oligomycin Injection (X3) FCCP Injection (X3) FCCP Injection (X3) Anti A + Rot Injection (X3)
Calibrate Mix Mix Mix Mix Mix
(04:00) (04:00) (04:00) (04:00) (04:00)
Equilibrate (12:00) Wait Wait Wait Wait Wait
(02:00) (02:00) (02:00) (02:00) (02:00)
Measure (03:00) Measure (03:00) Measure (03:00) Measure Measure (03:00)
(03:00)

Table 2: Protocol setup for bioenergetic profiling. Description of the steps and their length in minutes using the Seahorse Wave Desktop software. Abbreviations: FCCP = carbonyl cyanide-p-trifluoromethoxyphenylhydrazon; Rot = rotenone; Anti A = Antimycin A.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This paper describes the reproducible generation of human iPSC-derived brain organoids and their use for mitochondrial disease modeling. The protocol described here is modified based on a previously published work20. One major advantage of the present protocol is that it does not require the manual embedding of each organoid into a scaffolding matrix. In fact, the matrix solution is simply dissolved into the cell culture medium. Moreover, there is no need to employ expensive bioreactors, as organoids can be cultured in standard tissue culture 6-well plates positioned onto an orbital shaker inside the incubator. This procedure also enables the parallel cultivation of several plates containing different organoids derived from various individual lines, thereby increasing the throughput of the experiments and allowing the monitoring of potential differences emerging in the growth profiles of different organoids. We tested this protocol using different iPSCs derived from healthy controls and individuals affected by mitochondrial diseases, with consistent results.

For mitochondrial disease modeling, it is essential to use different markers to visualize the morphology and organization of the mitochondrial network. This procedure enables the investigation of whether mitochondrial number, morphology, or distribution might be altered in brain organoids derived from patients with mitochondrial diseases. The presence and organization of neural progenitors within the brain organoids could be of crucial importance for modeling mitochondrial disorders. We recently discovered that mutations causing the mitochondrial disease, Leigh syndrome, disrupt the cellular architecture and distribution of neural progenitor cells within patient-derived brain organoids18.

For performing bioenergetic profiling, we have adapted a method that was previously described for assessing the bioenergetics of pluripotent stem cells21. A recent protocol described how to carry out bioenergetic profiling of organoids derived from mouse small intestine, human colon, and colorectal tumors22. However, those organoids are quite small compared to brain organoids, and therefore, a different protocol, such as the one reported here, is needed for brain organoids. We recently employed this protocol for assessing the bioenergetic profile of human brain organoids carrying mutations in the surfeit locus protein 1 gene (SURF1) that causes the severe mitochondrial disease, Leigh syndrome18. We found that the OCR profile is particularly affected in Leigh syndrome organoids, as shown by a significant decrease in the basal OCR level, the ATP production rate, and the maximal respiration rate18.

In conclusion, we present here a detailed protocol for the robust generation of human brain organoids and describe how to perform experiments that would be important for the investigation of the disease mechanisms underlying mitochondrial diseases. Human brain organoids may also be of critical importance for elucidating the mitochondrial diversity in the human brain and its role in human health and diseases23. It is important to clarify that brain organoids generated with currently available protocols, including the one described here, still bear limitations. These include, for example, the lack of vascularization and the absence of microglia population24. These aspects need to be taken into consideration to interpret the results correctly.

For example, the lack of vasculature and microglia could limit compensatory mechanisms that may be in place in vivo. Patient-derived brain organoids might thus exhibit defects that are stronger than the ones observed in patients17,18. Moreover, despite a general reproducibility of this protocol20, line-to-line heterogeneity can be observed. To this end, when performing disease modeling studies, it is always important to systematically quantify the uniformity of control and patient organoids by assessing the patterns of morphology (size, layer) and the distribution of molecular markers across different organoids.

Finally, it is not possible to generate brain organoids from a single iPSC, limiting the feasibility of large-scale genetic screening with CRISPR/Cas9. Given the pace of research, it is likely that some of the current limitations of the protocol described here will soon be overcome. Optimized protocols will become available. These 3D models of mitochondrial diseases will hopefully enable the eventual discovery of implementable therapies for mitochondrial diseases, which are detrimental, and for incurable diseases with highly unmet medical needs.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors declare no competing financial or non-financial interests.

Acknowledgments

We thank Miriam Bünning for technical support. We acknowledge support from the Deutsche Forschungsgemeinschaft (DFG) (PR1527/5-1 to A.P.), Spark and Berlin Institute of Health (BIH) (BIH Validation Funds to A.P.), the United Mitochondrial Disease Foundation (UMDF) (Leigh Syndrome International Consortium Grant to A.P.), University Hospital Duesseldorf (Forschungskommission UKD to A.P.), and the German Federal Ministry of Education and Research (BMBF) (e:Bio young investigator grant AZ 031L0211 to A.P.).Work in the laboratory of C.R.R. was supported by the DFG (FOR 2795 "Synapses under stress", Ro 2327/13-1).

Materials

Name Company Catalog Number Comments
2-mercaptoethanol Gibco 31350-010
Affinity Designer Serif (Europe) Ltd Layout software; Vector graphics editor
Alexa Fluor 488 donkey anti-guinea pig Sigma Aldrich SAB4600033-250UL 1:300
Alexa Fluor 488 donkey anti-mouse Thermo Fisher Scientific A-31571 1:300
Antimycin A Sigma Aldrich 1397-94-0
Anti-β-Tubulin III (TUJ-1) Sigma Aldrich T8578 1:2000
Argon Laser Melles Griot Any other Laser, e.g., diode lasers emitting 488 is fine, too
Ascorbic acid Sigma A92902
B-27 with Vitamin A Gibco 17504044
Bacto Agar Becton Dickinson 3% in PBS, store solution at -20 °C
BDNF Miltenyi Biotec 130-093-0811
cAMP Sigma D0627
Cell Star cell culture 6 well plate Greiner-Bio-One 657160
Chemically Defined Lipid Concentrate Gibco 11905031
Confocal laser scanning microscope C1 Nikon Microscope Solutions Modular confocal microscope system
Corning Matrigel Growth Factor Reduced (GFR) Basement membrane matrix, Phenol Red-free, LDEV-free Corning 356231 Matrix component
CyQUANT Cell Proliferation Assay Kit Thermo Fisher C7026
DMEM/F12 ThermoFisher 31330038
DMSO Sigma D2660-100ML
Donkey anti-goat Cy3 Merck Millipore AP180C 1:300
Donkey anti-mouse Cy3 Merck Millipore AP192C 1:300
Donkey anti-rabbit Cy3 Merck Millipore AP182C 1:300
DPBS Gibco 14190250
DS-Q1Mc camera Nikon Microscope Solutions
Eclipse 90i upright widefield microscope Nikon Microscope Solutions
Eclipse E 600FN upright microscope Nikon Microscope Solutions
Eclipse Ts2 Inverted Microscope Nikon Microsope Solutions
EZ-C1 Silver Version 3.91 Nikon Microscope Solutions Imaging software for confocal microscope
FCCP Sigma Aldrich 370-86-5
Fetal Bovine Serum Gibco 10270-106
GDNF Miltenyi Biotec 130-096-291
Glasgow MEM Gibco 11710-035
Glass Pasteur pipette Brand 747715 Inverted
Glutamax Gibco 35050-061
Helium-Neon Laser Melles Griot Every other Laser, e.g., diode lasers emitting 594 is fine, too
Heparin Merck H3149-25KU
HERACell 240i CO2 Incubator Thermo Scientific 51026331
Hoechst 33342 Invitrogen H3570 1:2500
Image J 1.53c Wayne Rasband National Institute of Health Image processing Software
Injekt Solo 10 mL/ Luer Braun 4606108V
Knockout Serum Replacement Gibco 10828010
Laser (407 nm) Coherent Any other Laser, e.g., diode lasers emitting 407 is fine, too
Map2 Synaptic Systems No. 188004 1:1000
Maxisafe 2030i
MEM NEAA Gibco 11140-050
mTeSR Plus Stemcell Technology 85850 iPSC medium
Multifuge X3R Centrifuge Thermo Scientific 10325804
MycoAlert Mycoplasma Detection Kit Lonza # LT07-218
N2 Supplement Gibco 17502-048
Needle for single usage (23G x 1” TW) Neoject 10016
NIS-Elements Aadvanced Research 3.2 Nikon Imaging software
Oligomycin A Sigma Aldrich 75351
Orbital Shaker Heidolph Unimax 1010 Heidolph 543-12310-00
PAP Pen Sigma Z377821-1EA To draw hydrophobic barrier on slides.
Papain Dissociation System kit Worthington LK003150
Paraformaldehyde Merck 818715 4% in PBS, store solution at -20 °C
Pasteur pipette 7mL VWR 612-1681 Graduated up to 3 mL
Penicillin-Streptomycin Gibco 15140-122
Plan Apo VC 20x / 0.75 air DIC N2  ∞/0.17 WD 1.0 Nikon Microscope Solutions Dry Microscope Objective
Plan Apo VC 60x / 1.40 oil DIC N2 ∞/0.17 WD 0.13 Nikon Microscope Solutions Oil Immersion Microscope Objective
Polystyrene Petri dish (100 mm) Greiner Bio-One 664161
Polystyrene round-bottom tube with cell-strainer cap (5 mL) Falcon 352235
Potassium chloride Roth 6781.1
ProLong Glass Antifade Moutant Invitrogen P36980
Qualitative filter paper VWR 516-0813
Rock Inhibitior Merck SCM075
Rotenone Sigma 83-794
S100β Abcam Ab11178 1:600
SB-431542 Cayman Chemical Company 13031
Scalpel blades Heinz Herenz Hamburq 1110918
SMI312 Biolegend 837904 1:500
Sodium bicarbonate Merck/Sigma 31437-1kg-M
Sodium chloride Roth 3957
Sodium dihydrogen phosphate Applichem 131965
Sodium Pyruvate Gibco 11360070
SOX2 Santa Cruz Biotechnology Sc-17320 1:100
StemPro Accutase Cell Dissociation Reagent Gibco/StemPro A1110501 Reagent A
Super Glue Gel UHU 63261 adhesive gel
SuperFrost Plus VWR 631-0108
Syringe for single usage (1 mL) BD Plastipak 300015
TB2 Thermoblock Biometra
TC Plate 24 Well Sarstedt 83.3922
TC Plate 6 Well Sarstedt 83.392
TGFbeta3 Miltenyi Biotec 130-094-007
Tissue Culture Hood ThermoFisher 51032711
TOM20 Santa Cruz Biotechnology SC-11415 1:200
Triton-X Merck X100-5ML
UltraPure 0.5M EDTA Invitrogen 15575020
Vibratome Microm HM 650 V Thermo Scientific Production terminated, any other adjustable microtome is fine, too.
Vibratome Wilkinson Classic Razor Blade Wilkinson Sword 70517470
Whatman Benchkote Merck/Sigma 28418852
Wnt Antagonist I EMD Millipore Corp 3378738
XF 96 extracellular flux analyser Seahorse Bioscience 100737-101
XF Assay DMEM Medium Seahorse Bioscience 103680-100
XF Calibrant Solution Seahorse Bioscience 100840-000
XFe96 FluxPak (96-well microplate) Seahorse Bioscience 102416-100

DOWNLOAD MATERIALS LIST

References

  1. Koopman, W. J., Willems, P. H., Smeitink, J. A. Monogenic mitochondrial disorders. New England Journal of Medicine. 366 (12), 1132-1141 (2012).
  2. Gorman, G. S., et al. Mitochondrial diseases. Nature Review Disease Primers. 2, 16080 (2016).
  3. Vafai, S. B., Mootha, V. K. Mitochondrial disorders as windows into an ancient organelle. Nature. 491 (7424), 374-383 (2012).
  4. Carelli, V., Chan, D. C. Mitochondrial DNA: impacting central and peripheral nervous systems. Neuron. 84 (6), 1126-1142 (2014).
  5. Russell, O. M., Gorman, G. S., Lightowlers, R. N., Turnbull, D. M. Mitochondrial diseases: hope for the future. Cell. 181 (1), 168-188 (2020).
  6. Weissig, V. Drug development for the therapy of mitochondrial diseases. Trends in Molecular Medicine. 26 (1), 40-57 (2020).
  7. Tyynismaa, H., Suomalainen, A. Mouse models of mitochondrial DNA defects and their relevance for human disease. EMBO Reports. 10 (2), 137-143 (2009).
  8. Ma, H., et al. Metabolic rescue in pluripotent cells from patients with mtDNA disease. Nature. 524 (7564), 234-238 (2015).
  9. Galera-Monge, T., et al. Mitochondrial dysfunction and calcium dysregulation in Leigh syndrome induced pluripotent stem cell derived neurons. International Journal of Molecular Science. 21 (9), 3191 (2020).
  10. Zheng, X., et al. Alleviation of neuronal energy deficiency by mTOR inhibition as a treatment for mitochondria-related neurodegeneration. Elife. 5, 13378 (2016).
  11. Lorenz, C., et al. Human iPSC-derived neural progenitors are an effective drug discovery model for neurological mtDNA disorders. Cell Stem Cell. 20 (5), 659-674 (2017).
  12. Inak, G., et al. Concise review: induced pluripotent stem cell-based drug discovery for mitochondrial disease. Stem Cells. 35 (7), 1655-1662 (2017).
  13. Chiaradia, I., Lancaster, M. A. Brain organoids for the study of human neurobiology at the interface of in vitro and in vivo. Nature Neuroscience. 23 (12), 1496-1508 (2020).
  14. Lancaster, M. A., Knoblich, J. A. Generation of cerebral organoids from human pluripotent stem cells. Nature Protocol. 9 (10), 2329-2340 (2014).
  15. Liput, M., et al. Tools and approaches for analyzing the role of mitochondria in health, development and disease using human cerebral organoids. Developmental Neurobiology. , (2021).
  16. Winanto, K. Z. J., Soh, B. S., Fan, Y., Ng, S. Y. Organoid cultures of MELAS neural cells reveal hyperactive Notch signaling that impacts neurodevelopment. Cell Death and Disease. 11 (3), 182 (2020).
  17. Romero-Morales, A. I., et al. Human iPSC-derived cerebral organoids model features of Leigh Syndrome and reveal abnormal corticogenesis. bioRxiv. , (2020).
  18. Inak, G., et al. Defective metabolic programming impairs early neuronal morphogenesis in neural cultures and an organoid model of Leigh syndrome. Nature Communications. 12 (1), 1929 (2021).
  19. Falk, M. J. Neurodevelopmental manifestations of mitochondrial disease. Journal of Developmental & Behavioral Pediatrics. 31 (7), 610-621 (2010).
  20. Velasco, S., et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature. 570 (7762), 523-527 (2019).
  21. Pfiffer, V., Prigione, A. Assessing the bioenergetic profile of human pluripotent stem cells. Methods in Molecular Biology. 1264, 279-288 (2015).
  22. Ludikhuize, M. C., Meerlo, M., Burgering, B. M. T., Colman, R. M. J. Protocol to profile the bioenergetics of organoids using Seahorse. STAR Protocols. 2 (1), 100386 (2021).
  23. Menacho, C., Prigione, A. Tackling mitochondrial diversity in brain function: from animal models to human brain organoids. International Journal of Biochemestry & Cell Biology. 123, 105760 (2020).
  24. Del Dosso, A., Urenda, J. P., Nguyen, T., Quadrato, G. Upgrading the physiological relevance of human brain organoids. Neuron. 107 (6), 1014-1028 (2020).

Tags

Brain Organoids Mitochondrial Disease Modeling Reproducible Assessment Mitochondrial Properties Bioreactors Embedding Procedures Implementation Upscaling Interventional Targets IPSCs Feeder-free Conditions Culture Humidified Tissue Culture Incubator Carbon Dioxide Passage Enzyme-free Detachment Medium Cell Survival Rho-associated Protein Kinase Inhibitor Dissociate Cortical Differentiation Medium CDM1 Pre-warm PBS Reagent A
Generation of Human Brain Organoids for Mitochondrial Disease Modeling
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Le, S., Petersilie, L., Inak, G.,More

Le, S., Petersilie, L., Inak, G., Menacho-Pando, C., Kafitz, K. W., Rybak-Wolf, A., Rajewsky, N., Rose, C. R., Prigione, A. Generation of Human Brain Organoids for Mitochondrial Disease Modeling. J. Vis. Exp. (172), e62756, doi:10.3791/62756 (2021).

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