Summary

Efficient and Scalable Generation of Human Ventral Midbrain Astrocytes from Human-Induced Pluripotent Stem Cells

Published: October 02, 2021
doi:

Summary

Here, we present a method for reproducible generation of ventral midbrain patterned astrocytes from hiPSCs and protocols for their characterization to assess phenotype and function.

Abstract

In Parkinson’s disease, progressive dysfunction and degeneration of dopamine neurons in the ventral midbrain cause life-changing symptoms. Neuronal degeneration has diverse causes in Parkinson’s, including non-cell autonomous mechanisms mediated by astrocytes. Throughout the CNS, astrocytes are essential for neuronal survival and function, as they maintain metabolic homeostasis in the neural environment. Astrocytes interact with the immune cells of the CNS, microglia, to modulate neuroinflammation, which is observed from the earliest stages of Parkinson’s, and has a direct impact on the progression of its pathology. In diseases with a chronic neuroinflammatory element, including Parkinson’s, astrocytes acquire a neurotoxic phenotype, and thus enhance neurodegeneration. Consequently, astrocytes are a potential therapeutic target to slow or halt disease, but this will require a deeper understanding of their properties and roles in Parkinson’s. Accurate models of human ventral midbrain astrocytes for in vitro study are therefore urgently required.

We have developed a protocol to generate high purity cultures of ventral midbrain-specific astrocytes (vmAstros) from hiPSCs that can be used for Parkinson’s research. vmAstros can be routinely produced from multiple hiPSC lines, and express specific astrocytic and ventral midbrain markers. This protocol is scalable, and thus suitable for high-throughput applications, including for drug screening. Crucially, the hiPSC derived-vmAstros demonstrate immunomodulatory characteristics typical of their in vivo counterparts, enabling mechanistic studies of neuroinflammatory signaling in Parkinson’s.

Introduction

Parkinson's disease affects 2%-3% of people over 65 years of age, making it the most prevalent neurodegenerative movement disorder1. It is caused by degeneration of ventral midbrain dopamine neurons within the substantia nigra, resulting in debilitating motor symptoms, as well as frequent cognitive and psychiatric issues2. Parkinson's pathology is typified by aggregates of the protein, α-synuclein, which are toxic to neurons and result in their dysfunction and death1,2,3. As the dopaminergic neurons are the degenerating population in Parkinson's, they were historically the focus of research. However, it is apparent that another cell type in the brain, the astrocytes, also demonstrate abnormalities in Parkinson's, and are believed to contribute to degeneration in models of Parkinson's4,5,6,7.

Astrocytes are a heterogenous cell population that can transform both physically and functionally as required. They support neuronal function and health via a plethora of mechanisms, including the modulation of neuronal signaling, shaping of synaptic architecture, and trophic support of neuronal populations via secretion of specific factors6,8,9,10. However, astrocytes also have a substantial immunomodulatory role, integral to the development and propagation of neuroinflammation10,11. Neuroinflammation is observed in the brains of Parkinson’s patients, and significantly has recently been shown to pre-empt the onset of Parkinson's symptoms12,13,14,15, thereby taking the center stage in Parkinson's research.

At a cellular level, astrocytes are said to become reactive in response to injury, infection, or disease, as an attempt to facilitate neuroprotection9,6,10,16. Reactivity describes a shift in astrocyte phenotype characterized by changes in gene expression, secretome, morphology, and mechanisms of clearance of cell debris and toxic byproducts9,10,11,17. This reactive shift occurs in response to inductive signals from microglia, which are the immune cells of the CNS and the first responders to injury and disease9. Both astrocytes and microglia respond to inflammatory signals by moderating their own function and can transduce inflammatory signals and thus directly influence neuroinflammation9,10. However, the chronic nature of Parkinson's results in a transition where reactive astrocytes become toxic to neurons, and themselves promote degeneration and disease pathology6,9,10,18,19. Significantly it was recently demonstrated that blocking the transformation of astrocytes into the reactive neurotoxic phenotype prevents the progression of Parkinson's in animal models11. Astrocyte reactivity in the paradigm of neuroinflammation has therefore become a major focus of Parkinson's research, and similarly relates to a wide spectrum of diseases of the CNS. Together these findings build a picture of significant astrocytic involvement in the etiology of Parkinson's, emphasizing the need for accurate research models that recapitulate the phenotype of the human astrocyte populations that are involved in Parkinson's.

In the embryonic brain, neurons appear first, with the astroglial lineage, namely, the astrocytes and oligodendrocytes, appearing later in development6. In vivo and in vitro studies have highlighted a number of signaling pathways that appear to control the potency of neural progenitor cells from neuronal to astroglial derivatives. In particular, JAK/STAT, EGF, and BMP signaling play roles in the proliferation, differentiation, and maturation of astroglia20,21. These pathways have been the focus of in vitro protocols for the generation of astrocytes from pluripotent cells, including hiPSC6,22,23. There have been many successful examples of generating astrocytes from hiPSCs6,24,25. However, it is apparent that in vivo astrocytes in the CNS possess specific regional identities, which relate directly to their function, in accordance with the specific requirements of those astrocytes in relation to their specialized neuronal neighbors17,24,25,26. For example, relating specifically to the ventral midbrain, it has been demonstrated that astrocytes in this region express specific sets of proteins, including receptors for dopamine enabling communication with the local population of midbrain dopamine neurons26. Furthermore, ventral midbrain astrocytes demonstrate unique signaling properties26. Therefore, to study the role of ventral midbrain astrocytes in Parkinson's, we require an in vitro model that reflects their unique set of characteristics.

To address this, we have developed a protocol to generate ventral midbrain astrocytes (vmAstros) from hiPSCs. The resulting vmAstros exhibit characteristics of their in vivo ventral midbrain counterparts such as expression of specific proteins, as well as immunomodulatory functions. The results presented are from the differentiation of the NAS2 and AST23 hiPSC lines, which were derived and gifted to us by Dr. Tilo Kunath27. NAS2 was generated from a healthy control subject whereas AST23 is derived from a Parkinson's patient carrying a triplication in the locus encoding α-Synuclein (SNCA). These hiPSC lines have been previously characterized and used in a number of published research papers, including for the generation of various neural cell types27,28,29,30,31.

Protocol

1. Human hiPSC line thawing, maintenance, and cryopreservation For coating hiPSC culture plates, dilute vitronectin to 5 µg/mL (1:100) in PBS at 1 mL per 10 cm2 cell culture plate surface area. Leave for 1 h at room temperature. Remove vitronectin and proceed immediately to adding hiPSCs/media to the culture plate. NOTE When removing vitronectin from the plate, it is crucial that the culture surface is not allowed to dry out. To thaw hiPSCs, remove cryovials con…

Representative Results

Differentiation methodology and progression Here we present the details of both the methods employed for the generation of vmAstros and the protocols used for their subsequent phenotypic characterization. The method for generation of vmAstros is made up of several distinct differentiation stages, which can be monitored by microscopy and identifying distinct morphological characteristics (Figure 1A-F). A feeder-free hiPSC culture (<stro…

Discussion

This method for the generation of vmAstros from hiPSCs is highly efficient, generating pure cultures of vmAstros, and being reproducible for the generation of vmAstros from different hiPSC lines. This protocol was developed around the recapitulation of the developmental events required in the embryo to correctly pattern the developing midbrain and generate astrocytes and comprises three defined stages: 1) neural ventral midbrain induction to generate vmNPCs, 2) generation and expansion of vmAPCs, and finally 3) maturatio…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

This work was funded by a Parkinson's UK project grant (G-1402) and studentship. The authors gratefully acknowledge the Wolfson Bioimaging Facility for their support and assistance in this work.

Materials

Reagents
0.2M Tris-Cl (pH 8.5) n/a n/a Made up from Tris base and plus HCl
0.5M EDTA, PH 8 ThermoFisher  15575-020 1:1000 in D-PBS to 0.5 mM final 
1,4-diazabicylo[2.2.2]octane (DABCO) Sigma D27802-  25 mg/mL in Mowiol mounting solution
13 mm coverslips VWR 631-0149
2-Mercaptoethanol (50 mM) ThermoFisher 31350010
Accutase ThermoFisher 13151014
Advanced DMEM/F12 ThermoFisher 12634010 Has 1x NEAA but we add to final concentration of 2x (0.2 mM)
Ascorbic acid Sigma A5960 200 mM stock, 1:1000 to 200 µM final
B27 Supplement ThermoFisher 17504-044 50x stock
BSA Sigma 5470
Cell freezing media Sigma C2874 Cryostor CS10
Cell freezing vessel Nalgene 5100-0001
CHIR99021 Axon Medchem 1386 0.8 mM stock, 1:1000 dilution to 0.8 µM final
Cryovials  Sigma CLS430487
DAPI  Sigma D9542 1 mg/mL, 1:10,000 to 100ng/mL final (in PBS)
DMEM/F12 + Glutamax ThermoFisher 10565018
Dulbeccos-PBS (D-PBS without Mg or Ca) ThermoFisher 14190144 pH 7.2
E8 Flex medium kit ThermoFisher A2858501
Formaldehyde (36% solution) Sigma 47608
Geltrex ThermoFisher A1413302 1:100 or 1:400 in ice-cold DMEM/F12
Glutamax ThermoFisher 35050038 2 mM stock (1:200 in N2B27, 1:100 in ASTRO media to 20 µM final) 
Glycerol Sigma G5516
Human BDNF  Peprotech 450-02 20 µg/mL stock, 1:1000 to 20 ng/mL final
Human BMP4 Peprotech 120-05 20 µg/mL stock, 1:1000 to 20 ng/mL final
Human EGF Peprotech AF-100-15 20 µg/mL stock, 1:1000 to 20 ng/mL final
Human GDNF Peprotech 450-10 20 µg/mL stock, 1:1000 to 20 ng/mL final
Human insulin solution Sigma  I9278 10 mg/mL stock, 1:2000 to 5 µg/mL final
Human LIF Peprotech 300-05 20 µg/mL stock, 1:1000 to 20 ng/mL final
IL-6 ELISA kit Biotechne DY206
Isopropanol Sigma  I9516-4L For filling Mr Frosty cryostorage vessel
LDN193189 Sigma SML0559 100 µM stock, 1:10,000 dilution to 10 nM final
Mowiol 40-88 Sigma 324590
N2 Supplement ThermoFisher 17502048 100x stock
NEAA ThermoFisher 11140035 10 mM stock, 1:100 to 0.1 mM final 
Neurobasal media ThermoFisher 21103049
Normal Goat serum Vector Labs S-1000-20
Revitacell ThermoFisher A2644501 100x stock, 1:100 to 1x final
SB431542 Tocris 1614 10 mM stock, 1:1000 dilution to 10 µM final
SHH-C24ii Biotechne 1845-SH-025 200 µg/mL stock, 1:1000 to 200 ng/mL final
Tris-HCl Sigma  PHG0002 
Triton-X Sigma X100
Tween-20 Sigma  P7949
Vitronectin ThermoFisher A14700 1:50 in D-PBS
Antibodies for immunocytochemistry  Company Catalogue Number Host species
Antibody against S100b Sigma SAB4200671 Mouse; 1:200
Antibody against FOXA2 SCBT NB600501 Mouse; 1:50
Antibody against LMX1A ProSci 7087 Rabbit; 1:300
Antibody against LMX1A Millipore AB10533 Rabbit; 1:2000
Antibody against LMX1B Proteintech 18278-1-AP Rabbit; 1:300
Antibody against GLAST Proteintech 20785-1-AP Rabbit; 1:300
Antibody against GFAP Dako Z0334 Rabbit; 1:400
Antibody against CD49f Proteintech 27189-1-AP Rabbit; 1:100
Antibody against MSI1 Abcam ab52865 Rabbit; 1:400
Alexa Fluor 488 Goat Anti-Rabbit  ThermoFisher A32731 Goat; 1:500
Alexa Fluor 488 Goat Anti-Mouse ThermoFisher A32723 Goat; 1:500
Alexa Fluor 568 Goat Anti-Rabbit ThermoFisher A11036 Goat; 1:500
Alexa Fluor 488 Goat Anti-Mouse ThermoFisher A11031 Goat; 1:500

Referencias

  1. Poewe, W., et al. Parkinson disease. Nature Reviews Disease Primers. 3, 17013 (2017).
  2. Lees, A. J., Hardy, J., Revesz, T. Parkingson’s disease. Lancet. 373, 2055-2066 (2009).
  3. Braak, H., et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of Aging. 24, 197-211 (2003).
  4. Booth, H. D. E., Hirst, W. D., Wade-Martins, R. The role of astrocyte dysfunction in Parkinson’s disease pathogenesis. Trends in Neurosciences. 40, 358-370 (2017).
  5. Lindstrom, V., et al. Extensive uptake of alpha-synuclein oligomers in astrocytes results in sustained intracellular deposits and mitochondrial damage. Molecular and Cellular Neuroscience. 82, 143-156 (2017).
  6. Crompton, L. A., Cordero-Llana, O., Caldwell, M. A. Astrocytes in a dish: Using pluripotent stem cells to model neurodegenerative and neurodevelopmental disorders. Brain Pathology. 27, 530-544 (2017).
  7. di Domenico, A., et al. Patient-specific iPSC-derived astrocytes contribute to non-cell-autonomous neurodegeneration in Parkinson’s disease. Stem Cell Reports. 12, 213-229 (2019).
  8. Zhang, Y., Barres, B. A. Astrocyte heterogeneity: an underappreciated topic in neurobiology. Current Opinions in Neurobiology. 20, 588-594 (2010).
  9. Liddelow, S. A., et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 541, 481-487 (2017).
  10. Liddelow, S. A., Barres, B. A. Reactive astrocytes: production, function, and therapeutic Potential. Immunity. 46, 957-967 (2017).
  11. Yun, S. P., et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nature Medicine. 24, 931-938 (2018).
  12. Stokholm, M. G., et al. Assessment of neuroinflammation in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a case-control study. The Lancet Neurology. 16, 789-796 (2017).
  13. Williams-Gray, C. H., et al. Serum immune markers and disease progression in an incident Parkinson’s disease cohort (ICICLE-PD). Movement Disorders. 31, 995-1003 (2016).
  14. Gelders, G., Baekelandt, V., Vander Perren, A. Linking neuroinflammation and neurodegeneration in Parkinson’s disease. Journal of Immunology Research. 2018, 4784268 (2018).
  15. Hall, S., et al. Cerebrospinal fluid concentrations of inflammatory markers in Parkinson’s disease and atypical parkinsonian disorders. Scientific Reports. 8, 13276 (2018).
  16. Zamanian, J. L., et al. Genomic analysis of reactive astrogliosis. Journal of Neuroscience. 32, 6391-6410 (2012).
  17. Clarke, B. E., et al. Human stem cell-derived astrocytes exhibit region-specific heterogeneity in their secretory profiles. Brain. 143 (10), 85 (2020).
  18. Sevenich, L. Brain-resident microglia and blood-borne macrophages orchestrate central nervous system inflammation in neurodegenerative disorders and brain cancer. Frontiers in Immunology. 9, 697 (2018).
  19. Friedman, B. A., et al. Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Reports. 22, 832-847 (2018).
  20. Viti, J., Feathers, A., Phillips, J., Lillien, L. Epidermal growth factor receptors control competence to interpret leukemia inhibitory factor as an astrocyte inducer in developing cortex. Journal of Neuroscience. 23, 3385-3393 (2003).
  21. Nakashima, K., Yanagisawa, M., Arakawa, H., Taga, T. Astrocyte differentiation mediated by LIF in cooperation with BMP2. FEBS Letters. 457, 43-46 (1999).
  22. Serio, A., et al. Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proceedings of the National Academy of Sciences of the United States of America. 110, 4697-4702 (2013).
  23. Gupta, K., et al. Human embryonic stem cell derived astrocytes mediate non-cell-autonomous neuroprotection through endogenous and drug-induced mechanisms. Cell Death and Differentiation. 19, 779-787 (2012).
  24. Krencik, R., Ullian, E. M. A cellular star atlas: using astrocytes from human pluripotent stem cells for disease studies. Frontiers in Cellular Neuroscience. 7, 25 (2013).
  25. Krencik, R., Weick, J. P., Liu, Y., Zhang, Z. -. J., Zhang, S. -. C. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nature Biotechnology. 29, 528-534 (2011).
  26. Xin, W., et al. Ventral midbrain astrocytes display unique physiological features and sensitivity to dopamine D2 receptor signaling. Neuropsychopharmacology. 44, 344-355 (2019).
  27. Devine, M. J., et al. Parkinson’s disease induced pluripotent stem cells with triplication of the alpha-synuclein locus. Nature Communications. 2, 440 (2011).
  28. Chen, Y., et al. Engineering synucleinopathy-resistant human dopaminergic neurons by CRISPR-mediated deletion of the SNCA gene. European Journal of Neuroscience. 49, 510-524 (2019).
  29. Crompton, L. A., et al. non-adherent differentiation of human pluripotent stem cells to generate basal forebrain cholinergic neurons via hedgehog signaling. Stem Cell Research. 11, 1206-1221 (2013).
  30. Stathakos, P., et al. A monolayer hiPSC culture system for autophagy/mitophagy studies in human dopaminergic neurons. Autophagy. , 1-17 (2020).
  31. Stathakos, P., et al. Imaging autophagy in hiPSC-derived midbrain dopaminergic neuronal cultures for Parkinson’s disease research. Methods in Molecular Biology. 1880, 257-280 (2019).
  32. Bilican, B., et al. Mutant induced pluripotent stem cell lines recapitulate aspects of TDP-43 proteinopathies and reveal cell-specific vulnerability. Proceedings of the National Academy of Sciences of the United States of America. 109, 5803-5808 (2012).
  33. Cordero-Llana, O., et al. Clusterin secreted by astrocytes enhances neuronal differentiation from human neural precursor cells. Cell Death and Differentiation. 18, 907-913 (2011).
  34. Morrow, T., Song, M. R., Ghosh, A. Sequential specification of neurons and glia by developmentally regulated extracellular factors. Development. 128, 3585-3594 (2001).
  35. Namihira, M., et al. Committed neuronal precursors confer astrocytic potential on residual neural precursor cells. Developmental Cell. 16, 245-255 (2009).
  36. Ochiai, W., Yanagisawa, M., Takizawa, T., Nakashima, K., Taga, T. Astrocyte differentiation of fetal neuroepithelial cells involving cardiotrophin-1-induced activation of STAT3. Cytokine. 14, 264-271 (2001).
  37. Nakashima, K., Yanagisawa, M., Arakawa, H. Synergistic signaling in fetal brain by STAT3-Smad1 complex bridged by p300. Science. 284 (5413), 479-482 (1999).
  38. Nakashima, K., et al. Developmental requirement of gp130 signaling in neuronal survival and astrocyte differentiation. Journal of Neuroscience. 19, 5429-5434 (1999).
  39. Barbar, L., et al. CD49f is a novel marker of functional and reactive human iPSC-derived astrocytes. Neuron. 107, 436-453 (2020).
  40. Barbar, L., Rusielewicz, T., Zimmer, M., Kalpana, K., Fossati, V. Isolation of human CD49f(+) astrocytes and in vitro iPSC-based neurotoxicity assays. STAR Protocols. 1, 100-172 (2020).
  41. Gao, H. M., et al. Neuroinflammation and alpha-synuclein dysfunction potentiate each other, driving chronic progression of neurodegeneration in a mouse model of Parkinson’s disease. Environmental Health Perspectives. 119, 807-814 (2011).
  42. Gao, H. M., et al. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. Journal of Neuroscience. 28, 7687-7698 (2008).
  43. Horvath, I., et al. Co-aggregation of pro-inflammatory S100A9 with alpha-synuclein in Parkinson’s disease: ex vivo and in vitro studies. Journal of Neuroinflammation. 15, 172 (2018).
This article has been published
Video Coming Soon
Keep me updated:

.

Citar este artículo
Crompton, L. A., McComish, S. F., Stathakos, P., Cordero-Llana, O., Lane, J. D., Caldwell, M. A. Efficient and Scalable Generation of Human Ventral Midbrain Astrocytes from Human-Induced Pluripotent Stem Cells. J. Vis. Exp. (176), e62095, doi:10.3791/62095 (2021).

View Video