Method Article
Here we describe a protocol that facilitates the differentiation of human induced pluripotent stem cells into functional forebrain-specific astrocytes. This enables investigations into the role of glial cells in the pathogenesis of neurodevelopmental disorders, such as Fragile X Syndrome, and modeling of other brain disorders.
Fragile X syndrome (FXS), a leading inherited cause of autism spectrum disorder and intellectual disability, has been studied extensively using rodent models. More recently, human stem cell-derived model systems have also been used to gain mechanistic insights into the pathophysiology of FXS. However, these studies have focused almost exclusively on neurons. Further, despite growing evidence for a key role of glia in neuronal function in health and disease, little is known about how human astrocytes are affected by FXS.
Therefore, in this study, we successfully developed a protocol that captures key spatiotemporal milestones of brain development and aligns with the process of gliogenesis as well. Together this offers a useful framework for studying neurodevelopmental disorders. First, we patterned the human induced pluripotent stem cells into the neuroectodermal lineage with dual Suppressor of Mothers against Decapentaplegic (SMAD) inhibition and small molecules. Subsequently, we utilized specific growth factors and cytokines to generate control (CTRL) and FXS patient-derived astrocytic progenitor cells (APCs). Treatment of APCs with ciliary neurotrophic factor, a differentiating cytokine, regulated and drove the progenitor cells towards astrocytic maturation, yielding forebrain-specific glial fibrillary acidic protein-expressing astrocytes.
We found that these astrocytes are functional, as evidenced by their calcium responses to ATP application, and they exhibit dysregulated glycolytic and mitochondrial metabolism in FXS. Taken together, these findings provide a useful experimental platform of human origin for the investigation of cell-autonomous and non-cell-autonomous consequences of alterations in astrocytic function caused by neurodevelopmental disorders.
Fragile X Syndrome (FXS), a common inherited form of intellectual disability and autism spectrum disorder (ASD), is caused by the lack of fragile X messenger ribonucleoprotein (FMRP) produced by the fragile X messenger ribonucleoprotein 1 (FMR1) gene (OMIM: #300624, https://www.omim.org/entry/300624). FMRP plays a role in the regulation of mRNA translation, mRNA granule formation and transport, and microRNA-mediated regulation of gene expression1. Thus, loss of FMRP impacts not just brain development but also adult brain function. Both mRNA transcript levels of FMR1 and immunostaining for FMRP in the brain have shown high neuronal expression, alongside significant expression in glial cells as well2. However, a vast majority of earlier studies in animal models of FXS focused primarily on neurons and aberrations in their function. Consequently, little is known about the role of glia in FXS3. Traditionally thought of as "passive support" cells4, there is accumulating evidence for astrocytes being critical in mediating a wide range of neuronal functions5,6, including promoting synaptogenesis7, refinement of developing neural circuits8, and neurotransmitter recycling9. In parallel, there is growing evidence for the role of astrocytes in disease pathogenesis and many neurological conditions have been associated with astrocytic dysfunction10.
While much of the earlier work using animal models of FXS focused on identifying and validating various molecular targets in neurons for treating FXS, these preclinical findings have not always led to successful clinical outcomes. Further, setbacks in recent clinical trials also underscore the need for human-based model systems. Models of neurological disorders based on human stem cell-derived brain cells offer a powerful strategy to bridge this gap between mechanistic insights from animal studies and limited success with clinical outcomes for patients. However, only a handful of these studies have focused on astrocytes and that too mostly on astrocytes that were spinal in origin. This, in turn, is relevant in light of studies showing that the structure and function of astrocytes vary between brain regions11,12. Thus, a better understanding of disease-induced changes in human astrocytes also needs to take into account these brain region-specific differences in astrocytes. However, models of neurodevelopmental disorders using human stem cell-derived astrocytes that are specific to the forebrain remain comparatively underexplored13. Hence, to begin to address these gaps, we describe protocols for generating forebrain-specific astrocytes from patient-derived induced pluripotent stem cells (iPSCs) carrying FXS mutations; further, we show that astrocytes are functional and display altered metabolism.
All experiments using human induced pluripotent stem cells (hiPSCs) (Table 1) were performed after obtaining appropriate institutional regulatory approvals. Figure 1A represents complete differentiation protocol of hiPSCs to mature forebrain-specific astrocytes.
1. Maintenance and expansion of hiPSCs
2. Generation and characterization of astrocytic progenitor cells (APCs)
3. Generation and characterization of a homogeneous population of forebrain-specific astrocytes
Human induced pluripotent stem cell (hiPSC) colonies were maintained using commercially available defined medium and immunostained for pluripotency markers, Oct4 and Nanog (Figure 1B). FXS and CTRL APCs showed comparable highly enriched proportions of cells immunopositive for vimentin and NFIA (Figure 1C,D). We found that APCs derived from healthy and FXS hiPSCs showed substantial upregulation of FOXG1 compared to HOXB4 (Figure 1F) consistent with a predominant forebrain identity. We observed a significant reduction in the number of GFAP-expressing astrocytes in FXS groups compared to CTRL astrocytes without affecting the number of S100β-expressing astrocytes (Figure 2B). Western blot analysis showed that the astrocytes generated from both FXS lines lacked expression of FMRP (Figure 2C,D). Quantification of newly synthesized proteins did not reveal any significant difference between CTRL versus FXS lines in either APCs or astrocytes (Figure 3C). However, we found protein synthesis to be consistently higher in APCs than in their respective astrocytes for each line (Figure 3C). These results suggest a stage-specific reduction in de novo protein synthesis from APCs to astrocytes in both FXS and CTRL lines.
Control, as well as FXS astrocytes, exhibited ATP-induced calcium transients (Figure 4A). However, a more detailed analysis revealed several key differences between the individual calcium transients recorded from CTRL, FXS1, and FXS2 astrocytes (Figure 4B). We classified the astrocytes as responders and non-responders to ATP based on the presence or absence of a calcium response, respectively (Figure 4C). The number of responders in FXS1 and FXS2 was significantly lower than CTRL. Specifically, we observed a significant reduction in the peak amplitude of the first calcium transient evoked after ATP application in both FXS astrocyte lines (Figure 4D). Further, the total duration of the first calcium transient was significantly shorter in FXS1 and FXS2 lines than in CTRL (Figure 4E). However, the number of calcium events (quantified as the total number of events per 250 s; Figure 4F), was comparable between lines. Taken together, these findings reveal alterations in calcium responses elicited by ATP in hiPSC-derived FXS astrocytes. ECAR results suggest a higher rate of glycolysis, glycolytic capacity, and glycolytic reserve in hiPSC-derived FXS astrocytes (Figure 5A,B). We found no significant difference in basal respiration in CTRL and FXS astrocytes. Cellular ATP production showed no significant difference between CTRL and FXS astrocytes. FCCP stimulation elicited significantly lower maximal respiration in FXS astrocytes than in CTRL astrocytes (Figure 5C,D).
Figure 1: Derivation of forebrain-specific astrocytic progenitor cells from hiPSCs. (A) Illustrative workflow for generation of astrocytes from hiPSCs. (B) Representative images of hiPSCs displaying comparable expression of Oct4/Nanog in CTRL and FXS lines. (C) Homogeneous population of hiPSC-derived APCs expressed a similar number of Vimentin- and NFIA-positive cells. (D) No significant difference in the expression of Vimentin- and NFIA across CTRL and FXS APCs. Statistical analysis was done using two-factor ANOVA followed by Tukey's pairwise comparison. (E) Schematic illustrating region-specific expression of FOXG1 and HOXB4 during development in vivo. (F) Graphical representation of FOXG1 expression compared with HOXB4 (using qRT-PCR) across genotypes suggesting propensity towards forebrain lineage. Statistical analysis by two-factor ANOVA followed by Sidak's multiple comparison test. For all experiments N = 3 biological replicates. ***p < 0.001. Scale bar = 50 µm. Error bars represent SEM. Abbreviations: hiPSCs = human induced pluripotent stem cells; CTRL = Control; FXS = Fragile X Syndrome; APCs = astrocyte progenitor cells. Please click here to view a larger version of this figure.
Figure 2: Reduced number of glial fibrillary acidic protein-positive hiPSC-derived FXS astrocytes. (A) Representative images of hiPSC-derived astrocytes exhibiting S100β and GFAP expression. (B) Percentage of GFAP-positive astrocytes was significantly lower in FXS-derived astrocytes compared to CTRL, although both exhibited similar S100β expression. Statistical significance determined by two-factor ANOVA followed by Tukey's pairwise comparison. (C) Immunoblot representing expression of FMRP in astrocytes from CTRL and FXS. (D) Graphs showing absence of FMRP in hiPSC-derived FXS astrocytes. Statistical analysis done by single-factor ANOVA with Tukey's pairwise comparison. For all the above experiments N = 3 biological replicates. Scale bar = 50 µm. ***p < 0.001. Error bars represent SEM. Abbreviations: hiPSCs = human induced pluripotent stem cells; CTRL = Control; FXS = Fragile X Syndrome; GFAP = glial fibrillary acidic protein; FMRP = Fragile X messenger ribonucleoprotein. Please click here to view a larger version of this figure.
Figure 3: Reduced protein synthesis due to differentiation from APCs to astrocytes. (A) Schematic workflow depicting labeling and visualization of protein synthesis in hiPSC-derived APCs and astrocytes using FUNCAT metabolic labeling. (B) Representative images of (top) Vimentin-positive hiPSC-derived APCs and (bottom) GFAP-positive astrocytes with FUNCAT/human Nuclei Antibody (hNA) label in CTRL and FXS-derived cells. (C) FUNCAT/volume of hiPSC-derived APCs was significantly higher than corresponding derived astrocytes. For all experiments N = 3 biological replicates. Scale bar = 50 µm. **p < 0.01, ***p < 0.001. Whiskers represent 1.5 × IQR. Abbreviations: hiPSCs = human induced pluripotent stem cells; CTRL = Control; FXS = Fragile X Syndrome; GFAP = glial fibrillary acidic protein; FUNCAT = fluorescent non-canonical amino acid tagging. Please click here to view a larger version of this figure.
Figure 4: Deficient Ca2+ signaling evoked by ATP in hiPSC-derived FXS astrocytes. (A) Representative traces of Ca2+ transients recorded from individual astrocytes upon external application of ATP at 25th s. (B) Averaged F340/F380 ratios depicting Ca2+ transients over 250 s after ATP application. (C) Grouped data showing higher percentage of non-responders to ATP in hiPSC-derived FXS astrocytes. Statistical significance determined by two-factor ANOVA with Tukey's pairwise comparison. (D-F) Quantification of first peak response (amplitude and duration) and frequency of events. (D) Shows a significant reduction in amplitude. Statistical significance determined by single-factor ANOVA with Tukey's pairwise comparison and (E) duration in hiPSC-derived FXS astrocytes. Statistical significance determined by Kruskal-Wallis test with Dunn's multiple comparison test; data set represented as mean ranks with SEM. (F) Graphical representation of Ca2+ transient frequency in hiPSC-derived FXS astrocytes. Statistical analysis by single-factor ANOVA with Tukey's pairwise comparison. For all experiments N = 3 biological replicates; n = 19 cells for each cell line. *p < 0.05, **p < 0.01, ***p < 0.001. Error bars represent SEM. # = Number. Abbreviations: hiPSCs = human induced pluripotent stem cells; CTRL = Control; FXS = Fragile X Syndrome; GFAP = glial fibrillary acidic protein. Please click here to view a larger version of this figure.
Figure 5: Bioenergetic deficits in hiPSC-derived FXS astrocytes. (A) Line graph representing extracellular acidification rate of hiPSC-derived astrocytes (CTRL and FXS) after addition of 10 mM Glucose, 1 µM Oligomycin, and 50 mM 2-DG sequentially and plotted on a line graph. (B) Glycolysis, glycolytic capacity, and glycolytic reserve quantification showed an increase in hiPSC-derived FXS astrocytes. Statistical analysis done by two-factor ANOVA with Tukey's pairwise comparison. (C) Line graph depicting oxygen consumption rate measurement by sequential addition of 1.5 µM Oligomycin, 1 µM FCCP, and 0.5 µM antimycin A and Rotenone. (D) Basal respiration, maximal respiration, and ATP production were quantified from the line graph and hiPSC-derived FXS astrocytes showed significant decrease in maximal respiration in comparison to CTRL astrocytes. Statistical analysis done by two-factor ANOVA with Tukey's pairwise comparison. For all experiments N = 2 biological replicates. *p < 0.05, **p < 0.01, ***p < 0.001. Error bars represent SEM. Abbreviations: hiPSCs = human induced pluripotent stem cells; CTRL = Control; FXS = Fragile X Syndrome; ECAR = extracellular acidification rate; 2-DG = 2-deoxyglucose; OCR = oxygen consumption rate; FCCP = carbonyl cyanide-4 (trifluoromethoxy) phenylhydrazone. Please click here to view a larger version of this figure.
ID in manuscript | ID at source | Age | Sex | Reprogrammed cell line name | Reprogramming method | Starting cell type | G band karyotype |
(years) | |||||||
CTRL | ND30625 | 76 | M | CS25iCTR-18nxx | Episomal vectors | Fibroblast | Normal |
FXS1 | GM07072 | 22 | M | CS072iFXS-n4 | Episomal vectors | Fibroblast | Normal |
FXS2 | GM05848 | 4 | M | CS848iFXS-n5 | Episomal vectors | Fibroblast | Normal |
Table 1: Cell lines used in this studyto generate forebrain-specific astrocytes.
Supplemental Figure S1: Chromosomal analysis report. Normal GTG banding karyotype of (A) CTRL 46, XY, (B) FXS1 46, XY, and (C) FXS2 46, XY. Please click here to download this figure.
Supplemental Table S1: Primers used in this study for the characterization of cells. Please click here to download this table.
Here, we describe a method to generate human iPSC-derived astrocytes that serve as an assay platform for characterizing functional changes induced by FXS. These astrocytes are functionally viable in culture and exhibit various properties, as evidenced by a range of measurements carried out in the present study. A critical step in this protocol is the initial conversion of iPSCs to corticospheres using the enzymatic lifting method. At this stage, optimizing the incubation time for collagenase type IV and dispase is crucial. If this is not optimized, it may cause the iPSCs to disintegrate or form single cells, which will prevent the formation of corticospheres. These corticospheres are patterned towards forebrain specificity using dual SMAD inhibition15 and are further differentiated into astrocyte progenitor cells (APCs). To achieve a high population of APCs, the mechanical chopping of gliospheres is crucial. Real-time qPCR can be used to confirm the forebrain-specificity; we observed a significant fold change increase in FOXG1, a marker for the forebrain, compared to HOXB4, a marker for the spinal cord.
APCs are then differentiated into astrocytes using CNTF, an activator of the JAK-STAT signaling pathway16. Results from this study indicate that terminally differentiated astrocytes derived from FXS iPSCs have lower GFAP levels, which is suggestive of impaired maturity. Although our analyses did not find any difference in de novo protein synthesis in FXS astrocytes and APCs compared to controls, we found a reduction in de novo protein synthesis in the transition from APCs to astrocytes in both FXS and control lines. The significant lowering in translation between APCs and astrocytes is consistent with a more streamlined and curated translation that has been shown for neurons versus neural precursor cells17. Earlier work that has examined aberrant protein synthesis has utilized whole brain tissues, neurons, and patient-derived fibroblasts, not astrocytes or APCs18. The only published reports on protein synthesis from patient-derived lines are not in astrocytes, but from lymphoblastoid cells19 and fibroblasts20,21. In light of these variations across studies, future studies would benefit from using FUNCAT measurements in astrocytes derived from additional FXS-patient-derived lines.
We assessed the functionality of astrocytes by measuring intracellular calcium dynamics22 and bioenergetics. FXS astrocytes showed reduced peak amplitude and duration of ATP-induced calcium transients, with fewer cells responding to ATP when compared to control astrocytes. Together, these alterations suggest that FXS disrupts calcium homeostasis in human astrocytes. These results are consistent with an earlier study23,24 reporting impaired IP3 receptor activity in FXS fibroblasts. Further, we observed increased glycolysis, glycolytic capacity, and glycolytic reserve in FXS astrocytes, along with reduced mitochondrial oxygen consumption rates.
The present study describes the generation and characterization of a new in vitro model for FXS astrocytes by leveraging the power of patient-derived iPSCs. Notably, it is only through an earlier study using human astrocytes co-cultured with human neurons that we uncovered the critical role of astrocytes determining the electrophysiological phenotype of the neurons14. Those studies, however, focused almost entirely on disease-induced changes in neuronal activity, and nothing was explored in astrocytes. Here, this protocol offers a novel framework for shifting our focus to functional alterations in astrocytes. This, in turn, will shed light on previously unexplored astrocyte-neuron interactions, such as glutamate signaling and electrical responses. Our results at this stage emphasize the need to prioritize astrocytes in future research.
It is critical to recognize the limitations of this protocol. First, it involves mechanical chopping, which leads to DNA fragmentation as well. This can cause the chopped gliospheres to clump, eventually leading to cell death. Second, the seeding density of APCs during the terminal differentiation step to astrocytes is essential to prevent contact mediated cell-cell inhibition. It is recommended to optimize this across cell lines to prevent detachment from the surface of the culture vessel.
Here, our findings are the first such analysis in human astrocytes as all past studies were based on other cell types such as lymphoblastoid cell lines and human fibroblasts25. Dysfunctional energy metabolism is a known player in the etiology of autism spectrum disorders as shown by the increased levels of human plasma lactate, significantly lower oxygen consumption rate in granulocytes26, and reduced expression of mitochondrial oxidative phosphorylation genes in the anterior cingulated, motor cortex, thalamus, and cerebellum of children with ASD27. Similar changes in energy metabolism are seen in neurodegenerative disorders like Parkinson's disease28 and Alzheimer's disease29, suggesting that increased glycolysis may compensate for reduced functional function30. Future studies are needed to explore if similar mechanisms are in play in dysfunctional energy metabolism seen in FXS patient-derived astrocytes.
At present, generating forebrain-specific astrocytes is a significant achievement, but future advancements could allow for the creation of region-specific glia, such as cortical/hippocampal astrocytes. This would provide deeper insights into the distinct roles played by these astrocytes in regional brain functions, synaptic plasticity, and neuronal-glial interactions. The ability to generate such region-specific astrocytes could advance models of neurodevelopment and neurodegenerative diseases. Ultimately, this could lead to more precise therapeutic strategies targeting astrocyte dysfunction in specific brain regions.
The authors have no conflicts of interest to disclose.
We thank Prof. Sumantra Chattarji for intramural Funds. We thank Prof. Gaiti Hasan for access to the calcium imaging setup, Central Imaging and Flow Facility-National Centre for Biological Sciences, Padmanabh Singh and Prangya Hota for proofreading and suggestions, and the Labmate Asia team for their assistance in performing Seahorse XF assays.
Name | Company | Catalog Number | Comments |
1-Thioglycerol | Sigma-Aldrich | M6145 | |
Accutase solution | Sigma-Aldrich | A6964 | Enzyme cell detachment medium |
Adenosine 5′-triphosphate magnesium salt | Sigma-Aldrich | A9187 | |
Advanced DMEM/F-12 | ThermoFisher Scientific | 12634010 | |
Antibiotic-Antimycotic (100x) | ThermoFisher Scientific | 15240062 | |
B-27 Supplement (50x), serum-free | ThermoFisher Scientific | 17504044 | |
Bovine Serum Albumin | Sigma-Aldrich | A9418 | |
Chemically Defined Lipid Concentrate | ThermoFisher Scientific | 11905031 | |
Collagenase, Type IV, powder | ThermoFisher Scientific | 17104019 | |
Deoxyribonuclease I | Worthington Biochemical Corporation | LK003170 | |
Dimethyl sulfoxide | Sigma-Aldrich | D2650 | |
Dispase II, powder | ThermoFisher Scientific | 17105041 | |
DPBS, no calcium, no magnesium | ThermoFisher Scientific | 14190144 | |
Essential 8 Medium | ThermoFisher Scientific | A1517001 | |
FXS1, FXS2 | Coriell Institute of Medical Research | GM07072, GM05848 | FXS patient cells |
GlutaMAX Supplement | ThermoFisher Scientific | 35050061 | glutamine substitute |
Ham's F-12 Nutrient Mix | ThermoFisher Scientific | 11765054 | |
Healthy control cells | Cedars-Sinai Medical Center | ND30625 | healthy control cells |
Heparin sodium salt from porcine intestinal mucosa | Sigma-Aldrich | H3149 | |
IMDM | ThermoFisher Scientific | 12440053 | |
Insulin, human | Roche | 11376497001 | |
LDN 193189 | Stratech Scientific | S2618-SEL | |
Leukemia Inhibitory Factor human | Sigma-Aldrich | L5283 | |
Matrigel Growth Factor Reduced (GFR) Basement Membrane Matrix | Corning | 354230 | |
MEM Non-Essential Amino Acids Solution (100x) | ThermoFisher Scientific | 11140050 | |
Mouse FGF-basic (FGF-2/bFGF) Recombinant Protein | Peprotech | 450-33 | |
Mr. Frosty freezing container | ThermoFisher Scientific | 5100-0001 | cryobox |
N-2 Supplement (100x) | ThermoFisher Scientific | 17502048 | |
N-Acetyl-L-cysteine | Sigma-Aldrich | A9165 | |
Neurobasal Medium | ThermoFisher Scientific | 21103049 | |
Nunc Biobanking and Cell Culture Cryogenic Tubes | ThermoFisher Scientific | 377267 | |
Nunc Cell-Culture Treated 6 well dish | ThermoFisher Scientific | 140675 | |
Papain Dissociation System | Worthington Biochemical Corporation | LK003150 | |
Penicillin-Streptomycin | ThermoFisher Scientific | 15140122 | |
Recombinant Human CNTF Protein, CF | R&D Systems | 257-NT-010 | |
Recombinant Human EGF Protein, CF | R&D Systems | 236-EG-01M | |
RevitaCell Supplement (100x) | ThermoFisher Scientific | A2644501 | |
SB431542 | Tocris | 1614 | |
Seahorse XFe24 Analyzer | Agilent Technologies | ||
Seahorse XF Cell Mito Stress Test Kit | Agilent Technologies | 103015-100 | |
Seahorse XF Glycolysis Stress Test Kit | Agilent Technologies | 103020-100 | |
Tissue Culture Dishes-100 cm | Biostar Lifetech | TCD000100 | |
Transferrin | Roche | 10652202001 | |
VWR Razor Blades | VWR International | 55411-050 | |
Antibodies | |||
Primary antibody | Company | Catalog number | Dilution |
Oct4 (C-10) | Santa Cruz Biotechnology | sc-5279 | Dilution: 1:250 Secondary antibody: Goat anti-Mouse IgG, Alexa Fluor 568 |
Nanog | R & D Systems | AF1997 | Dilution: 1:100 Secondary antibody: Donkey anti-Goat IgG, Alexa Fluor 488 |
Vimentin | Abcam | Ab5733 | Dilution: 1:500 Secondary antibody: Goat anti-Chicken IgY, Alexa Fluor 488 |
NFIA | Abcam | Ab41851 | Dilution: 1:500 Secondary antibody: Goat anti-Rabbit IgG, Alexa Fluor 568 |
GFAP-cy3 | Sigma | C9205 | Dilution: 1:500 Secondary antibody: NA |
GFAP | DAKO | Z0334 | Dilution: 1:500 Secondary antibody: Goat anti-Rabbit IgG, Alexa Fluor 568 |
S100β | DAKO | IR504 | Dilution: 1:500 Secondary antibody: Goat anti-Rabbit IgG, Alexa Fluor 488 |
Anti-Nuclei Antibody, clone 235-1 | Merck Millipore | MAB1281 | Dilution: 1:1000 Secondary antibody: Goat anti-Mouse IgG1, Alexa Fluor 555 |
Secondary antibodies | Dilution | ||
Goat anti-Mouse IgG, Alexa Fluor 568 | Thermo Fisher Scientific | A11004 | 1:1000 |
Donkey anti-Goat IgG, Alexa Fluor 488 | Thermo Fisher Scientific | A11055 | 1:1000 |
Goat anti-Chicken IgY, Alexa Fluor 488 | Thermo Fisher Scientific | A11039 | 1:1000 |
Goat anti-Rabbit IgG, Alexa Fluor 568 | Thermo Fisher Scientific | A11011 | 1:1000 |
Goat anti-Rabbit IgG, Alexa Fluor 488 | Thermo Fisher Scientific | A11034 | 1:1000 |
Goat anti-Mouse IgG1, Alexa Fluor 555 | Thermo Fisher Scientific | A21127 | 1:1000 |
Request permission to reuse the text or figures of this JoVE article
Request PermissionThis article has been published
Video Coming Soon
Copyright © 2025 MyJoVE Corporation. All rights reserved