Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Neuroscience

Differentiation of Enteric Nervous System Lineages from Human Pluripotent Stem Cells

Published: May 17, 2024 doi: 10.3791/66133

Summary

Deriving enteric nervous system (ENS) lineages from human pluripotent stem cells (hPSC) provides a scalable source of cells to study ENS development and disease, and to use in regenerative medicine. Here, a detailed in vitro protocol to derive enteric neurons from hPSCs using chemically defined culture conditions is presented.

Abstract

The human enteric nervous system, ENS, is a large network of glial and neuronal cell types with remarkable neurotransmitter diversity. The ENS controls bowel motility, enzyme secretion, and nutrient absorption and interacts with the immune system and the gut microbiome. Consequently, developmental and acquired defects of the ENS are responsible for many human diseases and may contribute to symptoms of Parkinson's disease. Limitations in animal model systems and access to primary tissue pose significant experimental challenges in studies of the human ENS. Here, a detailed protocol is presented for effective in vitro derivation of the ENS lineages from human pluripotent stem cells, hPSC, using defined culture conditions. Our protocol begins with directed differentiation of hPSCs to enteric neural crest cells within 15 days and yields diverse subtypes of functional enteric neurons within 30 days. This platform provides a scalable resource for developmental studies, disease modeling, drug discovery, and regenerative applications.

Introduction

The enteric nervous system (ENS) is the largest component of the peripheral nervous system. The ENS contains more than 400 million neurons that are located within the GI tract and control nearly all functions of the gut1. Molecular understanding of the ENS development and function and its defects in enteric neuropathies requires access to a reliable and authentic source of enteric neurons. Access to human primary tissue is limited, and animal models fail to recapitulate key disease phenotypes in many enteric neuropathies. Human pluripotent stem cell (hPSC) technology has proven exceedingly beneficial in providing an unlimited source of desired cell types, especially those that are difficult to isolate from primary sources2,3,4,5,6,7. Here, we provide details of a stepwise and robust in vitro method to obtain ENS cultures from hPSCs. These scalable hPSC-derived cultures open avenues for developmental studies, disease modeling, and high-throughput drug screening and can provide transplantable cells for regenerative medicine.

Enteric neuron lineages are derived from neural crest (NC) cells following the ENS developmental path during embryogenesis. In embryos, NC cells emerge along the margins of the folding neural plate. They proliferate, migrate and give rise to many different cell types including sensory neurons, Schwann cells, melanocytes, craniofacial skeleton and enteric neurons and glia8,9,10. The cell fate decision depends on the distinct region along the anterior-posterior axis that the NC cells emerge from, i.e., cranial NC, vagal NC, trunk NC and sacral NC. The ENS develops from vagal and sacral NC cells with the former dominating the population of the enteric neurons owing to the extensive migration along the length of the bowel and colonizing the gut11.

Derivation of NC cells from PSCs commonly involves a combination of dual SMAD inhibition and WNT pathway activation12,13. Until recently, all NC induction protocols involved serum and other animal product additives in the culture conditions. Chemically undefined media not only lower the reproducibility of NC induction but also challenge mechanistic developmental studies. To overcome these challenges, Barber et al14 developed an NC induction protocol using chemically defined culture conditions and was hence advantageous over alternative methods that rely on serum-replacement factors (e.g., KSR)13,14. This was obtained by basal media replacements and optimizing a protocol originally presented by Fattahi et al to derive enteric NC cells from hPSCs. This improved system is the basis of the hPSC differentiation protocol that is presented here. It begins with induction of enteric neural crest (ENC) cells in a 15-day period by precise modulation of BMP, FGF, WNT and TGFβ signaling in addition to retinoic acid (RA). We then derive the ENS lineages by treating cells with glial cell line-derived neurotrophic factor (GDNF). All media compositions are chemically defined and yield robust enteric neuronal cultures within 30-40 days.

In addition to the monolayer cell culture system described here, alternative NC induction approaches have been developed which use free-floating embryoid-bodies15,16. Migratory cells in these cultures have been shown to express NC markers with a subset representing the vagal NC. Co-cultures with primary gut tissue have been used to enrich enteric NC precursors in these cultures. Media compositions in these studies contain a combination of different factors such as nerve growth factor, NT3 and brain-derived neurotrophic factor. At this stage, it is not fully clear how these factors might affect the enteric neuron precursor commitment identities. For an efficient comparison of the enteric NC induction in the monolayer and the embryoid-body-based cultures more data is required. Given the different culture layouts in these strategies, the use of each method should be considered and optimized according to specific application in mind.

The protocol presented here is reproducible and has been successfully tested by us and others using different hPSC (induced and embryonic) lines8,14,16.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. Media preparation

NOTE: Concentrations mentioned throughout the protocol are final concentrations of the media components. Prepare all media under sterile conditions in a laminar flow hood, and store at 4 °C in the dark. Use within 2 weeks.

  1. hPSC maintenance medium: Mix feeder-free hPSC maintenance medium supplement (20 µL/mL) with its base medium.
  2. Medium A: Add bone morphogenetic protein 4 (BMP4; 1 ng/mL), SB431542 (10 μM), and CHIR99021 (600 nM) to differentiation base medium.
  3. Medium B: Add SB431542 (10 μM), and CHIR99021 (1.5 μM) to differentiation base medium.
  4. Medium C: Add SB431542 (10 μM), CHIR99021 (1.5 μM) and RA (1 μM) to differentiation base medium.
  5. Neural crest complete (NCC) Medium: Add FGF2 (10 ng/mL), CHIR99021 (3 μM), N2 supplement (10 μL/mL), B27 supplement (20 μL/mL), L-glutamine supplement (10 μL/mL), and MEM NEAAs (10 μL/mL) to neurobasal medium.
  6. ENC Medium: Add GDNF (10 ng/mL), ascorbic acid (100 μM), N2 supplement (10 μL/mL), B27 supplement (20 μL/mL), L-glutamine supplement (10 μL/mL), and MEM NEAAs (10 μL/mL) to neurobasal medium.
  7. Ethylenediaminetetraacetic acid (EDTA) 1x: Dilute 500 mM EDTA (1000x) to a final concentration of 500 μM in PBS.
    NOTE: Use Ca2+ and Mg2+ free PBS throughout this protocol unless otherwise stated.

2. Coating of culture plates

  1. Basement membrane matrix coated plates: Quickly thaw, and dilute a 500 μL frozen aliquot of basement membrane matrix in 50 mL of chilled DMEM:F12 by vigorously pipetting using a 50 mL serological pipette. Coat the wells with the diluted basement membrane matrix solution (100 μL per cm2 of well surface area) and incubate them overnight at 37 °C. Aspirate the coating medium before use. To prevent gelatinization of basement membrane matrix, perform this step as quickly as possible and use cold DMEM:F12 for dissolving the frozen aliquot.
    NOTE: Basement membrane matrix temperature should be kept cold during thawing (on ice) and aliquoting (tubes on ice) to prevent coagulation.
  2. PO/FN/laminin coated plates: Prepare a 15 μg/mL polyornithine (PO) solution in PBS by diluting a 1000x stock. Coat wells with 100 μL of PO/PBS solution per cm2 of well surface area and incubate the plates overnight at 37 °C. The following day, aspirate PO/PBS and coat the wells at 100 μL per cm2 of well surface area with freshly made FN/laminin/PBS solution containing 2 μg/mL of fibronectin (FN) and 2 μg/mL of laminin. Plates can be used after a minimum of 2 h incubation at 37 °C. Aspirate FN/laminin/PBS before use.

3. Maintenance of hPSC culture

NOTE: All cell incubation steps are at 5% CO2 and at 37 °C in a humidified incubator.

  1. Aspirate hPSC medium from the hPSC culture and add fresh medium (200 μL per cm2 of well surface area) every other day until colonies are ~80% confluent.
  2. To passage, aspirate hPSC medium and wash cells with 100 μL of PBS per cm2 of well surface area.
    NOTE: It is important to use Ca2+ and Mg2+ free PBS to wash the cells.
  3. Add 500 μM EDTA (100 μL per cm2 of well surface area). Replace the lid of the plate. Watch through an inverted microscope and monitor the cells for detachment of colony edges (2-4 min).
  4. Use a 5 or 10 mL serological pipette to transfer the same volume of hPSC medium into each well and mechanically harvest the cells by pipetting up and down a few times.
    NOTE: Optimal EDTA incubation time depends on the iPSC line. Avoid vigorous pipetting for detaching colonies as it can lead to excessive colony dissociation and cell death. When passaging to start a differentiation plate, incubate cells with EDTA for 1-2 min longer.
  5. Transfer the cell suspension into a 15 mL conical tube. Spin the cells (2 min, 290 x g, 20-25 °C) and carefully discard the supernatant.
  6. Resuspend the cells in an appropriate volume of hPSC medium and transfer to wells of a new basement membrane matrix coated plate.
    NOTE: For regular hPSC maintenance use a 1:18 - 1:24 passaging ratio (1:18 means transferring one third of the cells dissociated from a single well of a 6-well plate into a full new plate). To start a differentiation plate, follow a 5:6 passaging ratio.
  7. Incubate at 5% CO2 and 37 °C.

4. Neural crest induction

NOTE: This step is schematically represented in Figure 1A. Begin NC differentiation when cells are about 80% confluent. This will be achieved within 1-2 days when cells are passaged at a 5:6 ratio (see above). Start NC differentiation on day 0 with pluripotent and fully undifferentiated hPSC cells. For a high efficiency, the colony borders should have minimal differentiating cells. Passage and plate hPSC for differentiation when colonies are large, or the center of the colony starts to thicken/darken when monitored using an inverted microscope. Paying attention to the confluency and morphology tends to be more reliable than the number of plated cells as this might vary depending on the cell line and can vary from 50,000-200,000 per cm2.

  1. On day 0, replace hPSC maintenance medium with Medium A containing 1 ng/mL BMP4, 10 μM SB431542, 600 nM CHIR99021 (200 μL of medium per cm2 of well surface area).
  2. On day 2, cells should be 100% confluent. Gently aspirate spent Medium A and feed cells with Medium B containing 10 μM SB431542, 1.5 μM CHIR99021 (200 μL of medium per cm2 of well surface area).
    NOTE: As cultures grow during the NC induction, cells may detach from the underlying monolayer. Avoid excess cell loss by gently removing old media and adding fresh media to the side of the well.
  3. On day 4, feed cells again with Medium B (200 μL of medium per cm2 of well surface area).
  4. On day 6, replace medium B with Medium C containing 10 μM SB431542, 1.5 μM CHIR99021 and 1 μM retinoic acid (400 μL of medium per cm2 of well surface area).
  5. On days 8 and 10, feed as day 6.

5. ENC spheroid formation

NOTE: This step is schematically represented in Figure 1B.

  1. On day 12, gently aspirate Medium C and wash cells with 100 μL of PBS per cm2 of well surface area. Add the same volume of cell detachment solution and incubate for 30 min at 5% CO2 and 37 °C.
  2. Dilute cell detachment solution by adding the same volume of NCC medium containing 10 ng/mL FGF2, 3 μM CHIR99021, 10 μL/mL N2 supplement, 20 μL/mL B27 supplement, 10 μL/mL L-glutamine supplement, and 10 μL/mL MEM NEAA. Using a serological pipette harvest the single cell suspension and add it to a 15 mL conical tube.
  3. Spin the cells (2 min, 290 x g, 20-25 °C) and carefully discard the supernatant.
  4. Using a 10 mL serological pipette, add 12 mL (for a full 6-well plate) of NCC medium and resuspend the cells completely by mechanically pipetting up and down 1-2 times. Transfer the cell suspension to an ultra-low attachment plate.
    NOTE: Approximately 10 cm2 of ENC monolayer cells are resuspended in 2 mL of NCC medium and transferred to one well of an ultra-low attachment plate. This equals to a full 6-well NC induction plate transferred into a full 6-well ultra-low attachment plate.
  5. On day 14, gently swirl the plates until small spheroids are gathered in the center of each well. Using a P1000 micropipette and in a slow circular motion, aspirate the spent NCC medium around the circumference of each well. Try to avoid removing the free-floating spheroids.
  6. Feed the cells with the original volume of NCC medium.

6. EN induction

NOTE: This step is schematically represented in Figure 1B.

  1. On day 15, apply the same technique used on day 14 to gently remove the medium and wash the spheroids with PBS. Remove as much PBS as possible avoiding the spheroids.
  2. Add cell detachment solution (100 μL per cm2 of well surface area) and dissociate the spheroids into single cells by incubating for 30 min at 5% CO2 and 37 °C.
  3. Using a 10 mL serological pipette, add an equal volume of ENC medium (containing 10 ng/mL GDNF, 100 μM ascorbic acid, 10 μL/mL N2 supplement, 20 μL/mL B27 supplement, 10 μL/mL L-glutamine supplement, and 10 μL/mL MEM NEAA in neurobasal medium) to each well and break the remaining spheroids by 2-3 rounds of mechanical pipetting. Transfer the cells to a 50 mL conical tube.
    NOTE: Avoid excessive sheer stress and cell death that can occur from forced pipetting or use of pipettes with smaller tip opening.
  4. Spin the cells (2 min, 290 x g, 20-25 °C) and carefully discard the supernatant. Using a 10 mL serological pipette, add ~ 5-10 mL of ENC medium and resuspend the cells completely by pipetting up and down 1-2 times.
  5. Count the concentration of viable cells using trypan blue and a cell counting method such as a hemocytometer.
    CAUTION: Trypan blue is a suspected carcinogen. Handle with care, dispose appropriately.
  6. Add enough volume of ENC medium aiming to plate approximately 300,000-400,000 viable cells per cm2 of surface area at a density of about ~ 1,000,000 cells per mL. Aspirate FN/laminin/PBS solution from wells of the plate.
    NOTE: Choose the plate format according to the assays that are planned for the EN cultures as this stage marks the final re-plating step of the protocol. Enteric neuron progenitors can be frozen on day 15. After dissociating the spheres with cell detachment solution, resuspend the progenitors at approximately 5 x 106 cells/mL in freezing medium such as 10% DMSO or Stem-cellbanker. When needed, thaw in warm ENC media with 5 μM Y-27632 ROCK inhibitor. Plate cells in the desired plate format. Replace the media with fresh ENC after a couple of hours.
  7. Gently add the cells into the PO/FN/laminin coated wells. Avoid bubbles getting trapped under the cell suspension preventing cells attachment to the PO/FN/laminin coating, especially when using plates with smaller well size such as 384-well plates. This could lead to cell death.
  8. Feed cells every other day with ENC medium (200 μL per cm2 of well surface area) until day 30-40, after which reduce the feeding frequency (to once or twice a week) but double the feeding volume (to 400 μL of medium per cm2 of well surface area).
    NOTE: This is important as excessive removing and adding media could facilitate detachment of neuronal culture from the surface of the wells. To prospectively prevent detachment especially for longer-term cultures, supplement ENC with FN (2 µg/mL) and laminin (2 µg/mL) once a week.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

This protocol provides a method to derive enteric neural crest and enteric neurons from hPSCs using chemically defined culture conditions (Figure 1A-B). Generating high-quality neurons depends on an efficient enteric neural crest induction step. This can be visually assessed by checking the morphology of the free-floating spheres that should look round with smooth surfaces with a size of approximately 0.1 - 0.4 mm as seen in Figure 1C. These spheroids should also express the neural crest marker SRY-Box transcription factor 10 (SOX10)17,18, as can be seen in Figure 1D. During enteric neuron induction phase, neurites can become visible as early as day 25-30 (Figure 1E). However, this can be delayed in differentiations with lower efficiency containing undesired mesenchymal cell contaminations. The human enteric nervous system is a collection of diverse neurons expressing different neurotransmitters. This heterogeneity is recapitulated in the in vitro protocol provided and the expression of key markers can be assessed by cell staining methods such as immunofluorescence imaging and flow cytometry according to previously described protocols13,14,19,20,21 (Figure 1F,G).

Figure 1
Figure 1: Deriving human ENC cells and enteric neuron subtypes from hPSCs. (A) Enteric neural crest (ENC) induction from hPSC. Protocol (days 0-12) showing media compositions for deriving ENC cells from hPSCs. (B) ENC spheroid formation and enteric neuron induction from ENC. Protocol (days 12-15) showing medium composition for ENC spheroid formation. Afterwards, enteric neurons are generated and maturated by treating with a medium containing GDNF and AA. (C) Representative phase contrast images of differentiating hPSCs at different time points of the protocol. Scale bar: 1000 µM. (D) SOX10::GFP expressing H9 iPSC-derived ENC spheroids on day 14. Scale bar: 1000 µm. (E) Phase contract image of day 33 neurons. Scale bar: 400 µm. (F-G) Immunofluorescence imaging (F) and flow-cytometry (G) of neurotransmitter markers in day 40 neurons. Scale bar: 50 µm. Abbreviations: BMP4 = recombinant human BMP4 protein; RA = retinoic acid; FGF2 = recombinant human FGF basic; GDNF = recombinant human glial cell line-derived neurotrophic factor; AA = ascorbic acid. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

The differentiation protocol described here provides a robust in vitro method to obtain enteric neurons from hPSCs within 30-40 days (Figure 1E) and enteric glia expressing glial fibrillary acidic protein, GFAP, and SOX10 in older cultures (> day 55)13,14,19,22. These neurons and glia are induced by stepwise differentiation of hPSCs into vagal and enteric neural crest cells followed by enteric neuron and glia progenitors13,14. The neural crest cells express neural crest markers, transcription factor AP-2 alpha and beta, TFAP2A and TFAP2B, respectively21. We previously showed that CD49 can be used as a marker to indicate the efficiency of hPSC-derived neural crest cells14. The efficiency and success of this method depends on a combination of factors. First, it is critical to perform all steps under sterile conditions. Testing hPSC cultures for mycoplasma contamination is important as it can adversely affect stem cell growth and is not detectable visually. Second, use of evenly and properly coated plates reduces the chance of patchy hPSC culture growth and detachment of NC cultures and older neurons from the surface of the wells. Third, for high efficiency in differentiation it is essential to start with pluripotent and fully undifferentiated hPSC cells. hPSC colonies should have minimal differentiating cells at the colony borders. hPSCs should be passaged or plated for differentiation when colonies are large, or the center of the colony starts to thicken/darken in color as observed through a phase contrast microscope. Undesired differentiated cells (usually mesenchymal cells), not only drop the overall differentiation efficiency, but they can result in detachment of neurons from the surface of the well as the cultures get older. It is therefore recommended to monitor the ENC cultures in the first 15 days carefully. Under efficient differentiation conditions dark disks appear on the surface of the culture ~day 6 which indicates efficient spheroid formation on days 12-15. In addition, in the spheroid phase (day 12-15) free-floating spheres should have smooth surfaces (Figure 1D). Maintaining the 3D spheroid phase on days 12-15 is generally sufficient to improve the purity of ENCs for the downstream differentiation of cultures into ENS lineages. However, for studies that require early and pure ENC progenitor cells populations, FACS using CD49D is recommended. CD49D is a specific surface marker for SOX10:hPSC-derived NC lineages and has been used successfully across cell lines13,14. For more information on isolating CD49D+ cells using FACS and recommendations on gating strategy readers are referred to the paper by Barber et al, Figure 2D, Box1 and Supplementary Figure 614. Fourth, to maximize efficiency, differentiation should start when hPSC cultures are monolayer and ~80% confluent. This means having a fully confluent culture on day 2. Lower confluency negatively affects the efficiency and very dense cultures tend to have a higher rate of cell death and can potentially lead to emergence of unwanted differentiated cells.

Re-plating cells on day 15 marks an important step in this protocol. This is the last cell transfer step for a monolayer neuronal culture and hence the plate layout should be chosen according to the assays planned. The number of cells transferred to each well can considerably affect the neuronal diversity and older neurons remaining attached to the surface of the wells. Generally, a cell density in the range of 250,000-350,000 cells per cm2 (about 75,000-100,000 cells per well of a 96-well plate or 500,000-700,000 per well of a 24-well plate) is desirable. Starting at day 30-40, one might consider adding fibronectin and laminin to the feeding medium (not more than once a week) to prospectively prevent detachment of neurons especially for cells planned for longer neuron cultures. At this point it is recommended to reduce the feeding frequency to once in every 5-7 days and leaving some medium in each well before adding extra fresh medium. This reduces the chance of applying unnecessary physical stress on the neuronal projections. Considering evaporation of medium (especially for wells in the border of the plate) one can remove 75 µL and add 100 µL of fresh medium to each well. Based on experience, cultures grown on polymer coverslip bottom imaging plates tend to live and stay attached to the surface of the well for a longer time. Therefore, considering the polymer coverslip bottom imaging plates for experiments requiring very old neuronal cultures such as co-cultures with other cell types is recommended. Although air-dried PO/FN/laminin coated plates might still yield in efficient differentiation, this has not been tested systematically for the purpose of this protocol. Hence, the use of freshly coated plates is recommended.

The protocol described here is based on a previously published protocol by Barber et al14. Compared to older ENC induction methods that use media containing serum-replacement factors (e.g., KSR)13,14, the current protocol media conditions are chemically defined. This is advantageous towards more consistent differentiations and allows more precise developmental studies by manipulating the culture conditions. This robust protocol reproducibly generates cultures of enteric neurons from independent induced and embryonic stem cell lines that have been tested, such as H9, UCSF4 and WTC11. A powerful aspect of the presented protocol is its capacity to generate a variety of ENS neuronal subtypes resembling those found in the human ENS. For example, expression of specific markers for different ENS neurochemical identities can be checked by flow cytometry and fluorescence imaging (Figure 1F,G). The typical range of hPSC-derived enteric neuronal populations positive for nitric oxide synthase 1 (NOS1) is 5%-20%; choline O-acetyltransferase (CHAT) is 40%-60%; gamma-aminobutyric acid (GABA) is 15%-30% and serotonin (5-HT) is 15%-30%. However, the extent to which the in vitro structural and functional connectivity between these subtypes accurately models the in vivo cellular networks requires further studies. In addition, single cell analyses to expand the transcriptional profiling of individual subtypes would not only help to better compare the in vivo and in vitro cellular identities but would also benefit developmental and molecular studies as well as future protocol optimizations towards enriching specific subtypes for drug discovery and regenerative medicine.

Despite the cultures containing different neuronal, and in older cultures glial cells, this protocol does not generate the other cell types naturally found in the in vivo niche of the ENS, such as muscle cells, epithelial cells, and immune cells. Studying the ENS interaction with these cell types in the context of health and disease requires investment in optimizing co-culture and intestinal organoids systems. Overall, a powerful method to generate different subtypes of the ENS in from hPSCs in vitro is described here. The scalable cultures are suitable for developmental studies of the human ENS, toxicology testing and disease modeling experiments aimed at developing drug therapies and providing transplantable cells for treating enteric neuropathies.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

The work was supported by grants from UCSF Program for Breakthrough Biomedical Research and Sandler Foundation, March of Dimes grant no. 1-FY18-394 and 1DP2NS116769-01, the NIH Director's New Innovator Award (DP2NS116769) to F.F. and the National Institute of Diabetes and Digestive and Kidney Diseases (R01DK121169) to F.F., H.M. is supported by Larry L. Hillblom Foundation postdoctoral fellowship, NIH T32-DK007418 fellowship and UCSF Program for Breakthrough Biomedical Research independent postdoctoral fellowship.

Materials

Name Company Catalog Number Comments
Ascorbic acid Sigma-Aldrich A5960
B27 supplement (serum free, minus vitamin A) Gibco 12587-010
Basement membrane matrix, Geltrex Gibco A14133-2
BMP4 R&D systems 314-BP
Cell culture centrifuge Eppendorf, model no. 5810R 02262501
Cell detachment solution, Accutase Stemcell Technologies 07920
CHIR99021 Tocris 4423
Conical tubes USA scientific 1475-0511, 1500-1211
Differentiation base medium, Essential 6 Life Technologies A1516401
DMEM/F-12 no glutamine Life Technologies 21331020
EDTA Corning MT-46034CI
Feeder-free hPSC maintenance medium, Essential 8 Flex Medium Kit Life Technologies A2858501
FGF2 R&D systems 233-FB/CF
Fibronectin Corning 356008
GDNF Peprotech 450-10
Hemocytometer Hausser Scientific 1475
Human pluripotent stem cells, H9 ESC WiCell RRID: CVCL_1240
Incubator with controlled humidity, temperature and CO2 Thermo Fisher Scientific Herralcell 150i
Inverted microscope Thermo Fisher Scientific EVOS FL
Laminar flow hood Thermo Fisher Scientific 1300 series class II, type A2
Laminin Cultrex 3400-010
L-glutamine supplement, Glutagro Corning 25-015-CI
MEM NEAAs Corning 25-025-CI
Multiwell plates, Falcon BD 353934, 353075
N-2 Supplement CTS A1370701
Neurobasal Medium Life Technologies 21103049
PBS (Ca and Mg free) Life Technologies 10010023
Pipette filler Eppendorf Z768715-1EA
Pipette tips USA scientific 1111-2830
Pipettes Fisherbrand 13-678-11E, 13-678-11F
PO Sigma-Aldrich P3655
polymer coverslip bottom imaging plates, ibidi ibidi 81156
RA Sigma-Aldrich R2625
SB431542 R&D systems 1614
Trypan blue stain, 0.4% Thermo Fisher Scientific 15250-061
Ultra-low attachment plates Fisher Scientific 07-200-601
Y-27632 dihydrochloride R&D systems 1254

DOWNLOAD MATERIALS LIST

References

  1. Gershon, M. D. The enteric nervous system: a second brain. Hosp Pract. 34 (7), 35-38 (1999).
  2. Haggarty, S. J., Silva, M. C., Cross, A., Brandon, N. J., Perlis, R. H. Advancing drug discovery for neuropsychiatric disorders using patient-specific stem cell models. Mol Cell Neurosci. 73, 104-115 (2016).
  3. Bose, A., Petsko, G. A., Studer, L. Induced pluripotent stem cells: a tool for modeling Parkinson's disease. Trends Neurosci. 45 (8), 608-620 (2022).
  4. Liu, C., Oikonomopoulos, A., Sayed, N., Wu, J. C. Modeling human diseases with induced pluripotent stem cells: from 2D to 3D and beyond. Dev Camb Engl. 145 (5), (2018).
  5. Marchetto, M. C. N., et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell. 143 (4), 527-539 (2010).
  6. Park, I. H., et al. Disease-specific induced pluripotent stem cells. Cell. 134 (5), 877-886 (2008).
  7. Lee, G., et al. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature. 461 (7262), 402-406 (2009).
  8. Shyamala, K., Yanduri, S., Girish, H. C., Murgod, S. Neural crest: The fourth germ layer. J Oral Maxillofac Pathol. 19 (2), 221-229 (2015).
  9. Crane, J. F., Trainor, P. A. Neural crest stem and progenitor cells. Annu Rev Cell Dev Biol. 22, 267-286 (2006).
  10. Thomas, S., et al. Human neural crest cells display molecular and phenotypic hallmarks of stem cells. Hum Mol Genet. 17 (21), 3411-3425 (2008).
  11. Heanue, T. A., Pachnis, V. Enteric nervous system development and Hirschsprung's disease: advances in genetic and stem cell studies. Nat Rev Neurosci. 8, 466-479 (2007).
  12. Mica, Y., Lee, G., Chambers, S. M., Tomishima, M. J., Studer, L. Modeling neural crest induction, melanocyte specification, and disease-related pigmentation defects in hESCs and patient-specific iPSCs. Cell Rep. 3 (4), 1140-1152 (2013).
  13. Fattahi, F., et al. Deriving human ENS lineages for cell therapy and drug discovery in Hirschsprung disease. Nature. 531 (7592), 105-109 (2016).
  14. Barber, K., Studer, L., Fattahi, F. Derivation of enteric neuron lineages from human pluripotent stem cells. Nat Protoc. 14 (4), 1261-1279 (2019).
  15. Li, W., et al. Characterization and transplantation of enteric neural crest cells from human induced pluripotent stem cells. Mol Psychiatry. 23 (3), 499-508 (2018).
  16. Workman, M. J., et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat Med. 23, 49-59 (2017).
  17. Southard-Smith, E. M., Kos, L., Pavan, W. J. Sox10 mutation disrupts neural crest development in Dom Hirschsprung mouse model. Nat Genet. 18 (1), 60-64 (1998).
  18. Lee, G., et al. Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells. Nat Biotechnol. 25 (12), 1468-1475 (2007).
  19. Majd, H., et al. hPSC-derived enteric ganglioids model human ENS development and function. BioRxiv. , (2022).
  20. Majd, H., et al. Deriving Schwann cells from hPSCs enables disease modeling and drug discovery for diabetic peripheral neuropathy. Cell Stem Cell. 30 (5), 632-647 (2023).
  21. Samuel, R. M., et al. Generation of Schwann cell derived melanocytes from hPSCs identifies pro-metastatic factors in melanoma. BioRxiv. , (2023).
  22. Richter, M. N., et al. Inhibition of muscarinic receptor signaling protects human enteric inhibitory neurons against platin chemotherapy toxicity. BioRxiv. , (2023).

Tags

human pluripotent stem cells hPSC neural crest NC enteric nervous system ENS enteric neuron enteric neuropathies in vitro differentiation developmental study disease modeling drug screening
This article has been published
Video Coming Soon
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Majd, H., Richter, M. N., Samuel, R. More

Majd, H., Richter, M. N., Samuel, R. M., Kalantari, A., Ramirez, J. T., Fattahi, F. Differentiation of Enteric Nervous System Lineages from Human Pluripotent Stem Cells. J. Vis. Exp. (207), e66133, doi:10.3791/66133 (2024).

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