Dario Besusso’s research while affiliated with National Institute of Molecular Genetics (INGM) and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (27)


Figure 1. Traditional methods for triplet repeats characterization. ( A ) Southern blotting requires genomic DNA digestion with restriction enzymes, f ollo w ed b y blotting and probing with a labeled DNA fragment that specifically h ybridiz es to the repeat containing region. ( B ) Fluorescence-PCR uses at least one fluorescent primer and performs fragment analysis using a capillary electrophoresis system. ( C ) Small-pool PCR relies on serial dilutions and multiple independent PCRs across the repeat, f ollo w ed b y electrophoresis and blotting. ( D ) Sanger sequencing of PCR amplicons, after allelic separation by electrophoresis, detects fluorescence emitted by chain-terminating nucleotides.
Figure 2. High-throughput sequencing methods for triplet repeat characterization. ( A ) PCR-based methods begin with PCR amplification of the region of interest; the resulting amplicons then undergo platform-specific library preparation for high-throughput sequencing. ( B ) CRISPR / Cas9-based enrichment methods in v olv e cutting DNA using the Cas9-CRISPR RNA s (crRNA s) comple x, f ollo w ed b y ligation of sequencing adapters to the free DNA ends. ( C ) In-silico -based enrichment methods (adaptive sampling or 'Read Until') are used with Oxford Nanopore Technologies (ONT) devices to selectively sequence DNA molecules. Based on the first sequenced bases, the voltage across the nanopore can be reversed to eject the molecule if it does not match an on-target region.
Figure 3. Proposed experimental strategies to characterize CAG repeats in HD. Based on the biological questions users may wish to address, a w orkflo w outlining the optimal experimental setup is proposed. An estimate of the costs for each approach is also provided: $ represents 10$; $$ represents 100$; $$$ represents 10 0 0$.
High-throughput sequencing platforms for triplet repeats characterization
Studies using sequencing-based approaches to characterize CAG repeats in HD
Navigating triplet repeats sequencing: concepts, methodological challenges and perspective for Huntington's disease
  • Literature Review
  • Full-text available

December 2024

·

27 Reads

·

1 Citation

Nucleic Acids Research

Simone Maestri

·

Davide Scalzo

·

·

[...]

·

Elena Cattaneo

The accurate characterization of triplet repeats, especially the overrepresented CAG repeats, is increasingly relevant for several reasons. First, germline expansion of CAG repeats above a gene-specific threshold causes multiple neurodegenerative disorders; for instance, Huntington’s disease (HD) is triggered by >36 CAG repeats in the huntingtin (HTT) gene. Second, extreme expansions up to 800 CAG repeats have been found in specific cell types affected by the disease. Third, synonymous single nucleotide variants within the CAG repeat stretch influence the age of disease onset. Thus, new sequencing-based protocols that profile both the length and the exact nucleotide sequence of triplet repeats are crucial. Various strategies to enrich the target gene over the background, along with sequencing platforms and bioinformatic pipelines, are under development. This review discusses the concepts, challenges, and methodological opportunities for analyzing triplet repeats, using HD as a case study. Starting with traditional approaches, we will explore how sequencing-based methods have evolved to meet increasing scientific demands. We will also highlight experimental and bioinformatic challenges, aiming to provide a guide for accurate triplet repeat characterization for diagnostic and therapeutic purposes.

Download

Figure 1. SI causes cell-type specific vulnerability. ( A ) Recent studies have shown that somatic expansions are not only tissue-specific, but also cell-type specific; ( B ) Vulnerable cell types preferentially undergo somatic expansion over the course of the patient's lifetime, ultimately leading to transcriptional dysregulation and cell death.
Figure 2. HTT allele str uct ures influence HD A O O. T he upper part of the diagram represents a reference HD allele with 42 CAG repeats f ollo w ed b y the typically human 'CAA-CAG' tract, leading to a protein with 42Q + 2Q (both CAA and CAG translate to glutamine, Q). The CCG-CCA pair [representing the initial tract of the proline-rich domain (PRD)] f ollo wing the CAGs is also shown. Middle and bottom sections: GWAS-identified variants in the HTT allele nucleotide sequence that alter A O O; specifically, (middle) the LOI disease haplotype, an A-to-G synonymous mutation in the polyQ tract, leads to the same protein as the reference HD allele (42Q + 2Q), but accelerates disease onset. Con v ersely (bottom), in the DUP disease haplotype, the inclusion of an additional 'CAA-CAG' tract dela y s disease onset despite adding two extra Qs to the protein (42Q + 4Q).
When repetita no-longer iuvant: somatic instability of the CAG triplet in Huntington's disease

December 2024

·

38 Reads

·

3 Citations

Nucleic Acids Research

Trinucleotide repeats in DNA exhibit a dual nature due to their inherent instability. While their rapid expansion can diversify gene expression during evolution, exceeding a certain threshold can lead to diseases such as Huntington’s disease (HD), a neurodegenerative condition, triggered by >36 C–A–G repeats in exon 1 of the Huntingtin gene. Notably, the discovery of somatic instability (SI) of the tract allows these mutations, inherited from an affected parent, to further expand throughout the patient’s lifetime, resulting in a mosaic brain with specific neurons exhibiting variable and often extreme CAG lengths, ultimately leading to their death. Genome-wide association studies have identified genetic variants—both cis and trans, including mismatch repair modifiers—that modulate SI, as shown in blood cells, and influence HD’s age of onset. This review will explore the evidence for SI in HD and its role in disease pathogenesis, as well as the therapeutic implications of these findings. We conclude by emphasizing the urgent need for reliable methods to quantify SI for diagnostic and prognostic purposes.



B001 CAGinSTEM, a human embryonic stem cell platform to identify genetic factors implicated in Huntington’s disease

September 2024

·

6 Reads

Journal of Neurology, Neurosurgery, and Psychiatry

Background It is well known that HD patients with similar CAG length show a wide range of variability in motor onset that can account for up to two decades. One possible explanation resides in the fact that the inherited CAG repeats may expand in somatic tissues, especially in post-mitotic neurons, giving rise to a HTT mosaicism that results in longer than inherited CAG tracts in affected tissues, such as the striatum and cortex. This expansion may continue during the lifetime of the individual and contribute to exacerbate neuronal toxicity and selective neuronal degeneration. More recently, trans- and cis- modifiers of age of onset (AOO) have been identified. However, if and how they cause the progressive accumulation of CAG instability is still unclear. Aim To identify new cis and trans modifiers of CAG instability, we aimed to establish an isogenic human stem cell platform that, combined with third generation long-read sequencing, allows to monitor HTT CAG size over time, both during mitotic cell replication and in post-mitotic neurons. Methods Starting from H9 human embryonic stem cell (hES) line, we inserted a monoallelic Recombinant Mediated Exchange Cassette within HTT exon 1, which can be subsequently exchanged with any exon1 variant in an efficient way. We generated a wide variety of exon 1 modified cell lines, which we refer to as the CAGinSTEM platform. Results Our data show that the CAGinSTEM platform is technically robust as for each genotype we have multiple cell lines which have been quality checked. By exploiting the properties of the CAGinSTEM platform, we are testing how CAG length and composition impact on CAG instability in terminally differentiated medium spiny neurons and in active proliferating hES cells. Conclusions The CAGinSTEM platform offers a distinctive biological model system designed to explore genotype-phenotype correlations and investigate the mechanisms underlying CAG instability accumulation in postmitotic human neurons and other cell types.Funded by an ERC Advanced Grant from the European Commission.


G007 Short tandem repeats sequencing: methodological challenges and perspective for Huntington’s disease

September 2024

·

6 Reads

Journal of Neurology, Neurosurgery, and Psychiatry

Background Huntington’s disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene encoding for the huntingtin protein. Recently, Genome Wide Association Studies (GWAS) have identified genomic variations, occurring both at the HTT locus and in genes mostly involved in mismatch repair pathways, to be associated with disease onset and progression. Such mutations correlate with modulations of somatic instability, which is now considered as the main driver of pathogenesis. Accordingly, the set-up of reliable sequencing-based methods for assessing somatic instability is of paramount importance. Aims In this work, we aimed to compare workflows for assessing somatic instability, by identifying strengths and drawbacks of each enrichment, sequencing and analysis method. Methods Based on research studies focusing on Short Tandem Repeats (STR) characterization available in the literature, we compared multiple enrichment methods coupled to various sequencing platforms and data analysis tools. We then performed a preliminary comparison on internal data, and discussed their accuracy in CAG sizing and sensitivity for rare alleles detection. Results We reported much higher enrichment for PCR-based methods, compared to CRISPR/Cas9 enrichment and adaptive sampling. Moreover, unlike long-read sequencing platforms, we described a decrease in sequencing quality at cycles above 300 for Illumina MiSeq. As a last point, we showed a preliminary comparison among sequencing-based methods on internal data, showing the impact of each step towards the most accurate STR characterization. Conclusions We anticipate that long-read sequencing of PCR amplicons incorporating UMIs may represent a valid alternative to PCR-free enrichment methods, providing highly accurate performances in terms of repeat length and rare alleles detection. Nonetheless, we recommend accurate optimization of PCR primers and conditions, as suboptimal number of PCR cycles may result in off-target products and inadequate number of PCR duplicates.


Neuroprotection by ADAM10 inhibition requires TrkB signaling in the Huntington’s disease hippocampus

August 2024

·

42 Reads

Cellular and Molecular Life Sciences

Synaptic dysfunction is an early pathogenic event leading to cognitive decline in Huntington’s disease (HD). We previously reported that the active ADAM10 level is increased in the HD cortex and striatum, causing excessive proteolysis of the synaptic cell adhesion protein N-Cadherin. Conversely, ADAM10 inhibition is neuroprotective and prevents cognitive decline in HD mice. Although the breakdown of cortico-striatal connection has been historically linked to cognitive deterioration in HD, dendritic spine loss and long-term potentiation (LTP) defects identified in the HD hippocampus are also thought to contribute to the cognitive symptoms of the disease. The aim of this study is to investigate the contribution of ADAM10 to spine pathology and LTP defects of the HD hippocampus. We provide evidence that active ADAM10 is increased in the hippocampus of two mouse models of HD, leading to extensive proteolysis of N-Cadherin, which has a widely recognized role in spine morphology and synaptic plasticity. Importantly, the conditional heterozygous deletion of ADAM10 in the forebrain of HD mice resulted in the recovery of spine loss and ultrastructural synaptic defects in CA1 pyramidal neurons. Meanwhile, normalization of the active ADAM10 level increased the pool of synaptic BDNF protein and activated ERK neuroprotective signaling in the HD hippocampus. We also show that the ADAM10 inhibitor GI254023X restored LTP defects and increased the density of mushroom spines enriched with GluA1-AMPA receptors in HD hippocampal neurons. Notably, we report that administration of the TrkB antagonist ANA12 to HD hippocampal neurons reduced the beneficial effect of GI254023X, indicating that the BDNF receptor TrkB contributes to mediate the neuroprotective activity exerted by ADAM10 inhibition in HD. Collectively, these findings indicate that ADAM10 inhibition coupled with TrkB signaling represents an efficacious strategy to prevent hippocampal synaptic plasticity defects and cognitive dysfunction in HD.


Telencephalic organoids present ventral and dorsal identities with impaired neuronal transcriptional signatures in HD
A Scheme of the protocol for telencephalic organoids. Created with BioRender.com. released under a Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International license. Abbreviations: BF=brightfield, IHC=immunohistochemistry, qPCR=quantitative PCR, scRNAseq=single cell RNA sequencing, TEM/SEM=transmission and scanning electron microscopy. B Brightfield images during differentiation. Scale bars = 250 µm C Immunohistochemistry analyses of telencephalic organoids during differentiation. At DIV 35 for ASCL1, GSX2, ISLT1 and NKX2.1; at DIV 60 for GAD67, CTIP2, GFAP, MAP2; at DIV 120 for GABA and TBR1. Scale bars = 100 µm D UMAP plots of scRNAseq analyses on CTRL (20CAG) and HD (56CAG) organoids at DIV 45 showing genotypes and clusters of subpopulations E UMAP plots for specific markers of telencephalic development (FOXG1) with progenitors (MKI67) and ventral identity (GSX2, ASCL1, DLX2, GAD2, DLX6) or dorsal identity (NEUROD6) F UMAP plots of scRNAseq analyses on CTRL (20CAG) and HD (56CAG) organoids at DIV 120 showing the genotypes and clusters of subpopulations G UMAP plots for specific markers of telencephalic development (FOXG1) with progenitors (MKI67) and ventral identity (GSX2, ASCL1, DLX2, GAD2) or dorsal identity (EOMES, NEUROD6) H Voxhunt heatmap of similarity between individual subpopulations of our organoids and human fetal brain by mapping onto BrainSpan human transcriptomic dataset I Venn diagram of DEGs between CTRLs and HD organoids showing which are in common between DIV 45, DIV 120, and KEGG pathway associated to HD (Wilcoxon test, two-sided, p < 0.05) J, K Volcano plot of p-value adjusted (p.adj.) for DEGs at DIV 45 or DIV 120, Wilcoxon test two-sided L Heatmap showing the results of gene ontology analysis of the common DEGs where the enriched GO terms are grouped according to their semantic similarity. (Biological Replicates for scRNAseq: At DIV 45 N=3 individual organoids per genotype and at DIV 120 N=a pool of 10 organoids from 2 cell lines per genotype. See fig. S1).
Self-organization and ventral maturation are altered in HD organoids
A UMAP and boxplot of score of DEGs of term “cortical cytoskeleton organization” upregulated in HD organoids at both time points. Abbreviations: AP=Apical Progenitors; BP=Basal Progenitors; IN=Inhibitory Neurons; CP=Cycling Progenitors; DP=Dorsal Progenitors; DN=Dorsal Neurons; VP=Ventral Progenitors; VN=Ventral Neurons B Scanning electron microscopy (SEM) images of CTRL and HD organoids at DIV 60. Scale bars = 5 µm C Diameter of neuronal processes (Dots shown are all the multiple processes measured in N = 6 organoids of independent biological replicates, but statistics is performed on the mean of processes per organoid. Dots coloured based on the 6 replicates). Unpaired two-tailed t-test, ****p<0,0001) D Immunohistochemistry analyses of CTRL (parental and 20CAG) and HD (48, 56 and 72 CAG) organoids at DIV 45-60 for GSX2, ASCL1, N-CADHERIN and PALS1. Scale bar 30 µm E Numbers of GSX2+ or ASCL1+ cells in each area (Manual counting. Anova One Way, Bonferroni post test, **p < 0,01, ****p < 0,0001. N ≥ 11 VZ-like structures, derived from 10 organoids of 2/3 biological replicates) F, G UMAP and boxplot of score of DEGs of terms “cerebral cortex GABAergic interneurons” and “subpallium development” downregulated in HD organoids H q-PCR for DLX2 and SP9 on CTRL and HD organoids at DIV 35, 60, and 120. (N = 5 independent biological replicates where each one is a pool of 4 organoids; error bars represent ± SEM; Anova One Way, Bonferroni post test, *p < 0,05; **p < 0,01) I Heatmap of gene expression of key dorso-ventral telencephalic markers in CTRL and HD organoids at DIV 120 J, K Immunohistochemistry analyses of CTRL and HD organoids at DIV 120 for GABA and TBR1. Scale bar 100 μm L Boxplots of the signature score of each subpopulation of organoids at DIV 120 and the bulk RNAseq from human fetal brain LGE and CTX (cortex) at 9pcw³⁹ (Wilcoxon test, two-sided, Bonferroni post test, ****p<0,0001). For A, F, G, and L the box plots show the median (centre line), upper and lower quartiles (box limits), and the highest and lowest values within 1.5× the IQR of the nearest hinge (whiskers). Source data are provided as a Source Data file.
Mosaic organoids reveal non-cell autonomous recovery of HD ventral identities
A Experimental scheme for the CTRL-HD mosaic organoids. Created with BioRender.com released under a Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International license B, C UMAP plots of scRNAseq analyses on mosaic organoids at DIV 120 for culture conditions (CTRL-mono, CTRL_co, HD_co, and HD_mono) and clusters of subpopulations identified with the Louvain algorithm (Ventral Progenitors, Ventral Neurons, Dorsal Progenitors, Dorsal Neurons) D Bubble plot of cell type proportions per condition E Cell density plots showing how the cells are concentrated in each condition. F Heatmap of number of DEGs (Wilcoxon test, two-sided, p < 0.05) between samples in a pairwise comparison G Heatmap of Jaccard similarity score, based on the proportion of the top 500 DEGS (defined by Wilcoxon rank-sum test) specifically identifying each condition (CTRL_mono, CTRL_co, HD_co, HD_mono) that are in common between two distinct conditions H Heatmap of Jaccard similarity score divided per subpopulations (Dorsal Neurons, Dorsal Progenitors, Ventral Progenitors, Ventral Neurons). I Venn diagram of DEGs in Ventral neurons revealing recovered genes by comparing the list of DEGs between CTRL organoids (CTRL_mono) and HD organoids (HD_mono) with a second list of DEGs between CTRL_mono and HD_co: the genes missing in this second list are the Recovered ones (in green), while genes included in both lists are the Unchanged one (in red) and genes that compare in the co-culture condition are new DEGs (in light blue) J Proportion between Recovered DEGs (in green) and Unchanged DEGs (in red) in each subpopulation K Top 30 GO terms associated to the recovered DEGs in ventral neurons, resulting from GO analysis performed with R package topGO (Fisher test, one-sided). L Automatic quantification of the area positive for GABA over GFP or TOM area (N=8 organoids from 2 independent biological replicates. error bars represent ± SEM. Anova One Way, Bonferroni post test. ****p < 0.0001, *p < 0.05). Source data are provided as a Source Data file.
HD cells recover maturation and fate determination when grown with CTRL cells
A Immunohistochemistry analyses of CTRL_mono (20GFP+20TOM) co-culture mosaic organoids (20GFP+56TOM and 20TOM+56GFP) and HD_mono (56GFP+56TOM) organoids at DIV 120 of differentiation for BASSOON and HOMER1. Scale bar 20 µm B Automatic quantification of numbers of synapses as BASSOON/HOMER1 co-localizing puncta in each condition. As it was not possible to quantify single GFP or TOM cells overlapping with the puncta staining, the quantification reports the total number of synapses in the co-culture mosaic condition (N = 8 organoids from 2 independent biological replicates. Data are normalized over the mean of CTRL values, error bars represent ± SEM. Anova One Way, Bonferroni post test. ***p < 0.001, **p < 0.01). C Differentiation trajectories of all mosaic organoids together, inferred with the method of RNA velocity D Length and confidence of the velocity vectors in each culture sample (Wilcoxon test, two-sided, Bonferroni post test, **p < 0,01; ****p < 0,0001) E, F Length and confidence of the velocity vectors in each individual subpopulation (Wilcoxon test, two-sided, Bonferroni post test, *p < 0,05; **p < 0,01; ***p < 0,001; ****p < 0,0001) G Pseudotime analysis in each culture sample performed using Monocle3 H, I Boxplots for the signature score (as described in methods) of comparison between our mosaic organoids over the bulk RNAseq from human fetal LGE and CTX (cortex) at 9pcw³⁹, focusing on individual subpopulation of each sample (Wilcoxon test, two-sided, Bonferroni post test, *p < 0,05; **p < 0,01; ***p < 0,001; ****p < 0,0001). For D, E, F, H, and I the box plots show the median (centre line), upper and lower quartiles (box limits), and the highest and lowest values within 1.5× the IQR of the nearest hinge (whiskers). For E, F and H, I exact p-values can be found in Supplementary Table 2 inside the Supplementary Information. Source data are provided as a Source Data file.
Cell-cell communications show cell type specific changes in mosaic organoids
A Experimental scheme for conditioned medium organoids with CTRL receiving media from HD and vice-versa. Created with BioRender.com released under a Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International license B q-PCR for DLX2, DLX6, GAD1, MAP2, and TBR2 on CTRL and HD canonical (mono) and organoids conditioned from HD (condit HD) or from CTRL (condit CTRL) at DIV 120. (N  = 4 pools of organoids from 2 independent biological replicates; error bars represent ± SEM; Anova One Way, Bonferroni post test, *p < 0,05; **p < 0,01) C CellChat circle plot communications between all subpopulations in each individual condition D CellChat bubble plot of gene expression for couples of ligands and receptors between cells grown together in the same organoid per each subpopulation. Abbreviations: VP=Ventral Progenitors; DP=Dorsal Progenitors; VN=Ventral Neurons; DN=Dorsal Neurons. Wilcoxon test, two-sided, Bonferroni adjustments for multiple comparisons E NeuronChat circle plot showing total communications between all subpopulations and conditions F NeuronChat stacked bar plot of gene expression for couples of ligands and receptors in each condition G Box plots of gene expression for GABRB3 receptor in ventral neurons and dorsal neurons for each condition. For G the box plots show the median (centre line), upper and lower quartiles (box limits), and the highest and lowest values within 1.5× the IQR of the nearest hinge (whiskers). Source data are provided as a Source Data file.
Huntington’s disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids

August 2024

·

106 Reads

·

2 Citations

Huntington’s disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.



hESC-derived striatal progenitors grafted into a Huntington’s disease rat model support long-term functional motor recovery by differentiating, self-organizing and connecting into the lesioned striatum

July 2023

·

153 Reads

·

9 Citations

Stem Cell Research & Therapy

Background Huntington’s disease (HD) is a motor and cognitive neurodegenerative disorder due to prominent loss of striatal medium spiny neurons (MSNs). Cell replacement using human embryonic stem cells (hESCs) derivatives may offer new therapeutic opportunities to replace degenerated neurons and repair damaged circuits. Methods With the aim to develop effective cell replacement for HD, we assessed the long-term therapeutic value of hESC-derived striatal progenitors by grafting the cells into the striatum of a preclinical model of HD [i.e., adult immunodeficient rats in which the striatum was lesioned by monolateral injection of quinolinic acid (QA)]. We examined the survival, maturation, self-organization and integration of the graft as well as its impact on lesion-dependent motor alterations up to 6 months post-graft. Moreover, we tested whether exposing a cohort of QA-lesioned animals to environmental enrichment (EE) could improve graft integration and function. Results Human striatal progenitors survived up to 6 months after transplantation and showed morphological and neurochemical features typical of human MSNs. Donor-derived interneurons were also detected. Grafts wired in both local and long-range striatal circuits, formed domains suggestive of distinct ganglionic eminence territories and displayed emerging striosome features. Moreover, over time grafts improved complex motor performances affected by QA. EE selectively increased cell differentiation into MSN phenotype and promoted host-to-graft connectivity. However, when combined to the graft, the EE paradigm used in this study was insufficient to produce an additive effect on task execution. Conclusions The data support the long-term therapeutic potential of ESC-derived human striatal progenitor grafts for the replacement of degenerated striatal neurons in HD and suggest that EE can effectively accelerate the maturation and promote the integration of human striatal cells.


In vitro-derived medium spiny neurons recapitulate human striatal development and complexity at single-cell resolution

December 2022

·

100 Reads

·

15 Citations

Cell Reports Methods

Stem cell engineering of striatal medium spiny neurons (MSNs) is a promising strategy to understand diseases affecting the striatum and for cell-replacement therapies in different neurological diseases. Protocols to generate cells from human pluripotent stem cells (PSCs) are scarce and how well they recapitulate the endogenous fetal cells remains poorly understood. We have developed a protocol that modulates cell seeding density and exposure to specific morphogens that generates authentic and functional D1- and D2-MSNs with a high degree of reproducibility in 25 days of differentiation. Single-cell RNA sequencing (scRNA-seq) shows that our cells can mimic the cell-fate acquisition steps observed in vivo in terms of cell type composition, gene expression, and signaling pathways. Finally, by modulating the midkine pathway we show that we can increase the yield of MSNs. We expect that this protocol will help decode pathogenesis factors in striatal diseases and eventually facilitate cell-replacement therapies for Huntington’s disease (HD).


Citations (16)


... In these studies, the precise measurement of CAG size and composition in individual brain cells, along with the corresponding transcriptional profiles, has become increasingly important. These aspects are discussed in detail in the accompanying article by some of the authors ( 17 ). Finally, we will review strategies aimed at reducing SI with the goal of fighting the disease. ...

Reference:

When repetita no-longer iuvant: somatic instability of the CAG triplet in Huntington's disease
Navigating triplet repeats sequencing: concepts, methodological challenges and perspective for Huntington's disease

Nucleic Acids Research

... Meera Purushottam meera.purushottam@gmail.com expansion disorders, especially in the brain, is a critical factor in disease biology [4]. Transcription-induced DNA slippage and instability may have profound biological consequences in repeat-associated neurodegenerative diseases, and account for expanded repeats in terminally differentiated cells like neurons [5]. ...

When repetita no-longer iuvant: somatic instability of the CAG triplet in Huntington's disease

Nucleic Acids Research

... Future strategies will likely focus on enhancing the potency, stability and duration of gene knockdowns in in vivo models, alongside developing high-throughput, long-term screening systems ( 58 ). Neuronal organoid cultures could serve as a valuable tool, enabling co-culturing and perturbation screening of various cis and trans -modifiers ( 59 ). Since HD phenotypes, such as SI, transcriptional dysregulation and mHTT aggregation formation ( 28 ,51 ) are often co-modulated, SI in HD-vulnerable cell types may serve as an effective early marker for potential therapies. ...

Huntington’s disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids

... In searching for an alternative donor cell source attention has fallen on the potential use of human-derived stem cells including human embryonic stem cells (hESCs) or human induced pluripotent stem cells (iPSC) [9]. Previous studies have demonstrated that hESC-derived neural stem cells (NSC) transplanted into the quinolinic acid (QA) lesion model of HD survive and generate regionspecific neurons expressing markers of MSNs [11][12][13][14][15]. To increase lineage specificity and enhance differentiation into MSNs, several groups differentiated hESCs into lateral ganglionic eminence precursors (LGEPs) [16][17][18][19][20]. ...

hESC-derived striatal progenitors grafted into a Huntington’s disease rat model support long-term functional motor recovery by differentiating, self-organizing and connecting into the lesioned striatum

Stem Cell Research & Therapy

... These transcriptomic findings were corroborated by abundant FOXG1 + and GSX2 + progenitors from 15DIV onward and BCL11B + , DARPP32 + , FOXP1 + , FOXP2 + MSNs from 24DIV (Fig 1I-L). We also found that striatal fate and MSN subtype neurogenesis were induced more rapidly in Activin A-treated iPSCs than SHHpatterned cultures (Fig 1G, S1J and S1K) 43 . ...

In vitro-derived medium spiny neurons recapitulate human striatal development and complexity at single-cell resolution

Cell Reports Methods

... Single-cell MACF1 expression data from 1/5-month undirected cerebral organoids were retrieved from the Human Organoid Single-Cell Browser. 15 Single-cell data of the adult human brain were obtained from the ScApeX database. 16 ...

Modeling human telencephalic development and autism-associated SHANK3 deficiency using organoids generated from single neural rosettes

... The expression and activity of ADAM10 play important roles in a few benign diseases like Alzheimer's disease, Parkinson's disease, thoracic aortic aneurysm, and Huntington's disease. (Fan et al., 2022;Vatanabe et al., 2021;Zhou et al., 2021;Zuccato et al., 2021). We focused on studying the mechanism of ADAM10 in malignant tumors. ...

A09 ADAM10 activity at the huntington’s disease presynapse
  • Citing Conference Paper
  • September 2021

Journal of Neurology, Neurosurgery, and Psychiatry

... At the progenitor cell level, Shi et al. [86] used single-cell RNA sequencing to study the diversity of human ganglionic eminences, identifying regional and temporal diversity among progenitors. Bocchi et al. [87] performed 96,789 single cells from the early human fetal striatum for Single-cell transcriptional analysis, which showed that D1 and D2 medium spiny neurons originated from the same progenitor cells and exhibited a continuum of fate determinants. Much research has also been carried out on single neuron de novo neuronization mechanisms. ...

The coding and long noncoding single-cell atlas of the developing human fetal striatum
  • Citing Article
  • May 2021

Science

... Due to limited availability of NVU-related cell types of a human origin, there is an increased demand of using human induced pluripotent stem cells (hiPSCs) to derive these cells. Importantly, the use of patient iPSCs provides a chance for the development of disease-specific models [134,135]. Therefore, the use of hiPSCs in combination with microfluidic technology, could contribute to a more comprehensive NVU modeling and advance our . CC-BY-NC-ND 4.0 International license made available under a (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. ...

hiPSCs for predictive modelling of neurodegenerative diseases: dreaming the possible
  • Citing Article
  • March 2021

Nature Reviews Neurology

... In contrast, the major calcium buffer in MFs, calbindin was unchanged suggesting importantly that calcium buffering is unaffected by the loss of ADAM10 (Fig. 4a). Similarly, the synaptic vesicle-associated protein VAMP1 was also not changed (Fig. 4a) confirming observations by Cozzolino et al. that presynaptic vesicle number is not affected in cKO ADAM10 mice [13]. We reasoned that the modest reduction in syt7 protein levels could not alone account for the profound loss of facilitation. ...

ADAM10 hyperactivation acts on piccolo to deplete synaptic vesicle stores in Huntington's disease
  • Citing Article
  • February 2021

Human Molecular Genetics